question
stringlengths 13
215
| ground_truth
stringlengths 2
3.15k
| context
stringlengths 0
157k
|
---|---|---|
Are synonymous sites in primates and rodents functionally constrained? | No. Synonymous sites in primates exhibited evidence for higher selective constraint that those in rodents. In primates up to 24% of synonymous sites could be under purifying selection, while in rodents synonymous sites evolved neutrally. | It has been claimed that synonymous sites in mammals are under selective
constraint. Furthermore, in many studies the selective constraint at such sites
in primates was claimed to be more stringent than that in rodents. Given the
larger effective population sizes in rodents than in primates, the theoretical
expectation is that selection in rodents would be more effective than that in
primates. To resolve this contradiction between expectations and observations,
we used processed pseudogenes as a model for strict neutral evolution, and
estimated selective constraint on synonymous sites using the rate of
substitution at pseudosynonymous and pseudononsynonymous sites in pseudogenes as
the neutral expectation. After controlling for the effects of GC content, our
results were similar to those from previous studies, i.e., synonymous sites in
primates exhibited evidence for higher selective constraint that those in
rodents. Specifically, our results indicated that in primates up to 24% of
synonymous sites could be under purifying selection, while in rodents synonymous
sites evolved neutrally. To further control for shifts in GC content, we
estimated selective constraint at fourfold degenerate sites using a maximum
parsimony approach. This allowed us to estimate selective constraint using
mutational patterns that cause a shift in GC content (GT ↔ TG, CT ↔ TC, GA ↔ AG,
and CA ↔ AC) and ones that do not (AT ↔ TA and CG ↔ GC). Using this approach, we
found that synonymous sites evolve neutrally in both primates and rodents.
Apparent deviations from neutrality were caused by a higher rate of C → A and C
→ T mutations in pseudogenes. Such differences are most likely caused by the
shift in GC content experienced by pseudogenes. We conclude that previous
estimates according to which 20-40% of synonymous sites in primates were under
selective constraint were most likely artifacts of the biased pattern of
mutation. |
Which animal bite can cause Capnocytophaga canimorsus infection? | Capnocytophaga canimorsus infection is typically associated with dog bites, especially in asplenic or immunocompromised patients, and typically manifest as sepsis and/or bacteremia. | CDC group DF-2 is the vernacular name given to a slow-growing gram-negative
bacterium that causes septicemia and meningitis in humans. Infections frequently
(one-third of cases) occur following dog bites or close contact with dogs or
occasionally with cats. Splenectomy and alcoholism appear to be strong
predisposing factors for DF-2 infection. In addition to 150 DF-2 strains
received for identification, we received 9 DF-2-like strains; 6 were isolated
from wound or eye infections, 3 of which were associated with dog bites and 1 of
which was associated with a cat scratch, and 3 were isolated from dog mouths.
The major characteristics of DF-2 include production of acid but no gas from
lactose and maltose and usually D-glucose; positive reactions for oxidase,
catalase, arginine dihydrolase, gliding motility, and
o-nitrophenyl-beta-D-galactopyranoside; growth enhanced by serum and by
incubation in a candle jar atmosphere; and negative reactions for sucrose,
raffinose, inulin, melibiose, nitrate reduction, indole, and growth on MacConkey
agar. DF-2-like strains had the same characteristics, except that acid was
formed from sucrose, raffinose, inulin, and melibiose. By the hydroxyapatite
method, DNAs from 12 DF-2 strains were 88% related in 60 degrees C reactions and
84% related in 75 degrees C reactions. Related sequences contained 0.5 to 1.5%
unpaired bases (divergence). Three DF-2-like strains were 73 to 80% related at
60 degrees C (with 2.0 to 2.5% divergence) and 68 to 75% related at 75 degrees
C. The relatedness of DF-2 and DF-2-like strains was 19 to 31% at 60 degrees
Celsius and 13 to 19% at 75 degrees Celsius. The relatedness of DF-2 and
DF-2-like strains to Capnocytophaga species was 4 to 7%. The DNA relatedness
date indicate that eh DF-2 and the DF-2-like strains are separate, previously
undescribed species. Both groups are phenotypically and genetically distinct
from Capnocytophaga species, although they do share several characteristics with
Capnocytophaga species, including cellular morphology, gliding motility,
cellular fatty acid composition, enhancement of growth in a candle jar
atmosphere, and G+C content. The new species differ from Capnocytophaga species
by their positive oxidase and catalase reactions. We chose to avoid creating a
new genus and proposed the names Capnocytophaga canimorsus sp. nov. for group
DF-2 and C. cynodegmi sp. nov. for the DF-2-like strains. Capnocytophaga canimorsus causes dog-bite wound induced sepsis in adults, but
infection may follow mucous membrane exposure. Systemic infection in children is
extremely rare. A neonate with frequent exposure to a family dog and no
cutaneous infection developed C. canimorsus meningitis. Suspicion of this
pathogen requires laboratory consultation. Parental counseling can limit the
risk of pet acquired infections. Capnocytophaga canimorsus, a commensal bacterium from dogs' mouths, can cause
septicemia or meningitis in humans through bites or scratches. Here, we describe
and characterize the inflammatory response of human and mouse macrophages on C.
canimorsus infection. Macrophages infected with 10 different strains failed to
release tumor necrosis factor (TNF)- alpha and interleukin (IL)-1 alpha .
Macrophages infected with live and heat-killed (HK) C. canimorsus 5 (Cc5), a
strain isolated from a patient with fatal septicemia, did not release IL-6,
IL-8, interferon- gamma , macrophage inflammatory protein-1 beta , and nitric
oxide (NO). This absence of a proinflammatory response was characterized by the
inability of Toll-like receptor (TLR) 4 to respond to Cc5. Moreover, live but
not HK Cc5 blocked the release of TNF- alpha and NO induced by HK Yersinia
enterocolitica. In addition, live Cc5 down-regulated the expression of TLR4 and
dephosphorylated p38 mitogen-activated protein kinase. These results highlight
passive and active mechanisms of immune evasion by C. canimorsus, which may
explain its capacity to escape from the host immune system. Capnocytophaga canimorsus has been recognized as an opportunistic pathogen
causing systemic infections in immunocompromised individuals. It is part of the
normal oral flora of the dog, and can be responsible for localized wound
infections in humans in consequence of bites. This microorganism causes also
septicemia, meningitis, endocarditis, ocular infections and rarely brain
abscess. We describe the case of an immunocompetent 28-year-old male with
temporal brain abscess from Capnocytophaga canimorsus secondary to dog's bite. Capnocytophaga canimorsus is a commensal bacterium from the canine oral flora,
which can cause septicemia or meningitis in humans upon bite wound infections.
C. canimorsus 5 (Cc5), a strain isolated from a patient with fatal septicemia,
was used to investigate the interaction between C. canimorsus and J774.1 mouse
macrophages. J774.1 cells infected at high multiplicity with Cc5 did not
phagocytose nor kill Cc5 within 120 min of infection, unless the bacteria were
opsonized with specific antibodies. Opsonization with complement, however, did
not increase phagocytosis. Moreover, infection of J774.1 cells with live Cc5 led
to the release of a soluble factor, which interfered with the ability of
macrophages to kill other phagocytosed bacteria. These results provide an
example of how C. canimorsus neutralizes the innate immune system. BACKGROUND: Dog bites are the most common animal bite injuries occurring in the
United States. Estimated infection rates range between 15% and 20%.
Polymicrobial infections are most common. Capnocytophaga canimorsus (C.
canimorsus) is a Gram-negative rod strongly associated with dog bites, and is
known to cause life-threatening infection in humans.
OBJECTIVES: 1) Outline epidemiology of dog bites in the United States; 2)
Identify host factors associated with infection, and common pathogens; 3)
Discuss microbiology of C. canimorsus; 4) Discuss common clinical manifestations
of C. canimorsus infection; 5) Outline treatment options.
CASE REPORT: A 42-year-old woman with a remote history of Hodgkin's lymphoma
(treated with irradiation) and thyroid carcinoma, both of which were in
remission, presented to the Emergency Department with fever, abdominal pain, and
diarrhea. She was found to be in septic shock. She was aggressively resuscitated
and administered broad-spectrum antibiotics. Blood cultures grew C. canimorsus
in 2/4 bottles. The patient recalled being bitten by the family dog 48 h before
her initial presentation. She made an uneventful recovery. She was felt to be
"functionally hyposplenic" due to her prior irradiation.
CONCLUSIONS: C. canimorsus is a rare pathogen strongly associated with dog
bites. By eliciting a history of animal bite, clinicians may be able to alert
the laboratory of suspected C. canimorsus infection. Prolonged laboratory
incubation times may be necessary as the organism is fastidious. Predisposing
conditions include, among others, prior splenectomy and alcoholism. The
mortality rate from C. canimorsus sepsis is high, so treatment should be
promptly initiated. Sudden and unexpected nontraumatic death in individuals with asplenia or
hyposplenia is usually due to fulmit bacterial sepsis, most often involving
Streptococcus pneumoniae, Neisseria meningitidis, and Hemophilus influenzae. We
report a case of a previously well 40-year-old man who died 5 hours after
hospital admission. At autopsy Waterhouse-Friderichsen syndrome was identified
and Capnocytophaga canimorsus was subsequently isolated on antemortem blood
cultures. Infection of humans with this organism is most often due to dog bite
or contact. Upon specific inquiry it was ascertained that 2 days before
admission the deceased had suffered a superficial bite to his hand by his pet
Staffordshire Bullterrier dog. His relevant history included a previous
splenectomy following blunt abdominal trauma. Asplenia and hyposplenia at
autopsy should prompt microbiological testing with consideration of unusual
organisms such as C. canimorsus. Although histories of animal contact or injury
are often not available at the time of autopsy, this should also be considered
in cases of apparent fulmit sepsis. In individuals with asplenia or
hyposplenia, dog bites do not have to involve excessive tissue trauma, vascular
compromise, or blood loss to be lethal. Described in this study is the case of a 53-year-old woman who developed a
life-threatening infection caused by the bacterium Capnocytophaga canimorsus (C.
canimorsus), subsequent to being bitten by a dog. The patient presented to an
Emergency Department with a 24-h history of diarrhoea and vomiting with
dehydration but within 36 h of presentation developed an overwhelming severe
sepsis with septic shock, disseminated intravascular coagulation, acute renal
failure, metabolic acidosis and threatened acute respiratory failure requiring
urgent intensive care intervention. At subsequent questioning her husband
volunteered that she had been bitten on the wrist by the family dog 24h prior to
the onset of symptoms; this bite had been extremely minor, requiring no
treatment at the time and leaving only a very superficial wound. The causative
organism was finally identified two weeks later as C. canimorsus, a common
commensal in the oral flora of dogs. C. canimorsus has been reported as a rare
cause of severe infection in susceptible individuals; however this case is of
particular interest as there were no apparent predisposing factors conferring
risk of severe infection. This case also raised significant practice issues for
the treating hospital. Capnocytophaga canimorsus is a gram-negative bacterial species hosted in the
oral cavity of dogs. C. canimorsus can cause sepsis, meningitis and
endocarditis. Penicillin is the drug of choice. However, the species is a
slow-grower and sometimes missed in blood cultures. Patients with a history of
alcoholism, splenectomy or immunodeficiency are at an increased risk of
contracting serious infections with C. canimorsus following dog bites. We report
a case story of C. canimorsus meningitis contracted after a dog bite. BACKGROUND: Animal bites are typically harmless, but in rare cases infections
introduced by such bites can be fatal. Capnocytophaga canimorsus, found in the
normal oral flora of dogs, has the potential to cause conditions ranging from
minor cellulitis to fatal sepsis. The tendency of C. canimorsus infections to
present with varied symptoms, the organism's fastidious nature, and difficulty
of culturing make this a challenging diagnosis. Rarely, bacterial cytotoxins
such as those produced by C. canimorsus may act as causative agents of TTP,
further complicating the diagnosis. Early recognition is crucial for survival,
and the variability of presentation must be appreciated. We present the first
known case of C. canimorsus infection resulting in TTP that initially presented
as splenic infarction.
CASE PRESENTATION: 72-year-old Caucasian male presented with a four-day history
of abdominal pain, nausea, vomiting, diarrhea, and intermittent confusion. On
presentation, vital signs were stable and the patient was afebrile. Physical
examination was unremarkable apart from petechiae on the inner left thigh, and
extreme diffuse abdominal pain to palpation and percussion along with positive
rebound tenderness. Initial investigations revealed leukocytosis with left shift
and thrombocytopenia, but normal liver enzymes, cardiac enzymes, lipase, INR and
PTT. Abdominal CT demonstrated a non-enhancing spleen and hemoperitoneum,
suggesting complete splenic infarction. Although the patient remained afebrile,
he continued deteriorating over the next two days with worsening
thrombocytopenia. After becoming febrile, he developed microangiopathic
hemolytic anemia and hemodynamic instability, and soon after was intubated due
to hypoxic respiratory failure and decreased consciousness. Plasma exchange was
initiated but subsequently stopped when positive blood cultures grew a
gram-negative organism. The patient progressively improved following therapy
with piperacillin-tazobactam, which was switched to imipenem, then meropenem
when Capnocytophaga was identified.
CONCLUSIONS: There is a common misconception amongst practitioners that the
presence of systemic infection excludes the possibility of TTP and vice versa.
This case emphasizes that TTP may occur secondary to a systemic infection,
thereby allowing the two processes to coexist. It is important to maintain a
wide differential when considering the diagnosis of either TTP or C. canimorsus
infection since delays in treatment may have fatal consequences. Capnocytophaga canimorsus is part of normal gingival flora of dogs and cats. The
organism can cause septicemia, meningitis, and endocarditis in humans after
contact with dogs or cats. In spite of the frequency of gastrointestinal
symptoms in C. canimorsus infection patients, specific gastrointestinal disease
or clinical images have not been reported. We report a case of C. canimorsus
bacteremia presenting with acute cholecystitis in elderly woman. She suffered
from general fatigue and right upper abdominal pain. She had leukocytosis and
abnormal liver function tests. She showed abnormal findings of the gallbladder
by abdominal computed tomography and ultrasonography. She was diagnosed with
acute cholecystitis without gallstones and was administered with antibiotics.
C. canimorsus was isolated from blood cultures. A history of an insignificant
wound secondary to a dog bite was elicited. She recovered completely with
antibiotic treatment. This case revealed that C. canimorsus bacteremia can be
presented with acute cholecystitis, suggesting that C. canimorsus could cause
cholecystitis. And this cholecystitis can be treated with antibiotics without
operation. Physicians seeing patients with acute cholecysitis should ask
questions regarding animal contact. Newly named in 1989, Capnocytophaga canimorsus is a bacterial pathogen found in
the saliva of healthy dogs and cats, and is transmitted to humans principally by
dog bites. This review compiled all laboratory-confirmed cases, animal sources,
and virulence attributes to describe its epidemiology, clinical features, and
pathogenesis. An estimated 484 patients with a median age of 55 years were
reported, two-thirds of which were male. The case-fatality rate was about 26%.
Its clinical presentations included severe sepsis and fatal septic shock,
gangrene of the digits or extremities, high-grade bacteremia, meningitis,
endocarditis, and eye infections. Predispositions were prior splenectomy in 59
patients and alcoholism in 58 patients. Dog bites before illness occurred in
60%; additionally, in 27%, there were scratches, licking, or other contact with
dogs or cats. Patients with meningitis showed more advanced ages, higher male
preponderance, lower mortality, and longer incubation periods after dog bites
than patients with sepsis (p < 0.05). Patients with prior splenectomy presented
more frequently with high-grade bacteremia than patients with intact spleens
(p < 0.05). The organism possesses virulence attributes of catalase and
sialidase production, gliding motility, cytotoxin production, and resistance to
killing by serum complement due to its unique lipopolysaccharide. Penicillin is
the drug of choice, but some practitioners prefer third-generation
cephalosporins or beta-lactamase inhibitor combinations. C. canimorsus has
emerged as a leading cause of sepsis, particularly post-splenectomy sepsis, and
meningitis after dog bites. Animal bites represent a significant global health problem and account for
approximately 1-2% of all visits to the emergency department. The vast majority
of animal bite injuries are inflicted by dogs (80-90%,) and cats (5-15%). The
most common complication following an animal bite is a wound infection, which
tends to be polymicrobial and include both aerobic and anaerobic bacteria mainly
of oropharyngeal origin. The likelihood of a cat bite becoming infected is
double of that of a dog bite. Pasteurella spp. predominates in infected dog and
cat bites. Dog bite injuries can be also associated with Capnocytophaga
canimorsus, an aggressive organism which can cause disseminated infections
(sepsis) and death, particularly in immunocompromised individuals. Early
aggressive local wound cleansing is the most important therapy to prevent
infection after animal bites. Due to the polymicrobial etiology of infected bite
wounds, broad-spectrum antibiotics, covering both aerobic and anerobic bacteria,
are often recommended as empiric treatment of animal bites. We describe a case of a life-threatening septicemia resulting from a previous
dog bite wound. The isolated bacterium was Capnocytophaga canimorsus, a
slow-growing Gram-negative bacillus commonly found in dog saliva. Known risk
factors for invasive C. canimorsus infections are alcohol abuse, cigarette
smoking, splenectomy or other forms of immunosuppression. Any clinician seeing
patients with a history of a dog bite should consider this pathogen as a
causative agent and take detailed history regarding exposure to animals. C. canimorsus and C. cynodegmi are dog and cat commensals which can be
transmitted to humans via bites or scratches and can cause sepsis, meningitis,
endocarditis, and eye- or wound infections. Recently an additional
Capnocytophaga species was identified as part of the oral flora of healthy dogs
and was given the name "C. canis". We previously identified a Capnocytophaga
isolate that could not be typed with available diagnostic tests including
MALDI-TOF, 16S rRNA sequencing or species-specific PCR. This strain and 21 other
Capnocytophaga spp isolated in Sweden from clinical blood- or wound-cultures
were subjected to whole genome sequencing using the Illumina platform.
Phylogenetic analysis revealed that the previously non-typable isolate belongs
to the putative new species "C. canis". Since this strain was isolated from a
wound it also shows that members of "C. canis" have the potential to be
pathogenic. In addition, our phylogenetic analysis uncovered an additional
species of Capnocytophaga, which can be transmitted from dogs and cats to
humans, suggesting a speciation within the Capnocytophaga family that has not
been observed before. We propose the name of "C. stomatis" for this putative
novel species. Capnocytophaga canimorsus is a gram-negative, capnophilic rod constituting
normal bacterial flora of the oral cavity of dogs and cats. It is also
considered to be an etiological factor of infections in human that may lead to
multiple complications, i.a. sepsis, endocarditis and meningitis. C. canimorsus
poses a serious threat, especially to patients with asplenia, cirrhosis or
alcohol abuse. In most cases, infection occurs after a dog bite. Isolation and
identification of the bacteria from the biological material is difficult and
often delayed because of slow growth of the bacteria on microbiological media.
Gold standard for bacteriological identification of C. canimorsus is polymerase
chain reaction method. Amoxicillin with clavulanic acid is considered the drug
of choice used in prophylaxis of C. canimorsus infections. Based on the data
available from the literature, the authors present the epidemiology, risk
factors, clinical picture, diagnostic methods and treatment of the C. canimorsus
infection. BACKGROUND: Capnocytophaga canimorsus is a bacterium of the normal oral flora of
dogs and cats. Human infection is caused by animal bite but is rarely observed,
mainly in immunocompromised patients. We present 2 cases of C. canimorsus
infection that occurred in immunocompetent patients and caused multiorgan
failure and in both cases severe neurologic involvement.
CASE REPORT: In the first case, we present a 69-year-old immunocompetent woman
with septic shock derived from skin and soft tissue infection after a dog's
bite. She developed ischemic necrosis evolving to gangrene of both forefeet and
hands, infective aortic endocarditis, and neurologic involvement caused by large
hemispheric hypodense lesions compatible with ischemic septical lesions. In the
second case, we present a 65-year-old immunocompetent man with meningitis after
a dog's bite. Despite antibiotic therapy, he developed neurologic clinical
deterioration, with right sensitive hemisyndrome associated with lack of
strength and motor skills of the right hand. Radiologic findings were consistent
with the diagnosis of cerebritis. WHY SHOULD AN EMERGENCY PHYSICIAN BE AWARE OF
THIS?: Clinicians should always be aware of this pathogen, both in
immunocompromised and immunocompetent patients, and consider prophylactic
antibiotics after exposure. INTRODUCTION: Capnocytophaga canimorsus infections are associated with dog
bites, especially in asplenic or immunocompromised patients, and typically
manifest as sepsis and/or bacteremia. Meningitis has been rarely described, and
its diagnosis may be delayed due to poor or slow growth using traditional
culture techniques. We provide our experience using polymerase chain reaction
(PCR) to establish the diagnosis and perform a comprehensive review of C.
canimorsus meningitis cases to provide summary data on the clinical
manifestations, diagnosis, and outcomes of this unusual infection.
METHODS: A systematic review of the peer-reviewed English literature (PubMed,
Embase, Ovid Medline) from January 1966 to March 2018 was conducted to identify
cases of C. canimorsus meningitis. Data collected included demographics, risk
factors, cerebrospinal fluid (CSF) findings, PCR results, treatments, and
outcomes. Descriptive statistics are presented as numbers (percentages) and
medians (ranges).
RESULTS: A total of 37 patients were reviewed with a median age of 63 years
(12 days to 83 years) with a male predomice (76%). A relatively low
proportion had an immunocompromised state (16% splenectomy and 5% steroid use);
the most common risk factor was alcoholism (19%). Fifty-nine percent reported a
dog bite (all within ≤ 14 days prior to presentation), while 22% reported a
non-bite dog exposure, 3% reported cat bite, and 3% reported both dog and cat
exposures; 11% reported no animal contact. CSF parameters included a median
white count of 1024 cells/mm3, 81% had neutrophilic predomice, median protein
of 190 mg/dl, and median glucose CSF/serum ratio 0.23. In 54% of cases, blood
cultures were positive for C. canimorsus (median, 4 days) and 70% had positive
CSF cultures (median, 5 days). PCR established the diagnosis in eight (22%)
cases. Antibiotic therapy was given for a median of 15 days (range, 7 to
42 days). Prognosis was overall favorable with only one (3%) death reported and
adverse neurologic and/or physical sequelae in 19% of the survivors.
CONCLUSION: C. canimorsus meningitis is a rare but increasingly important
clinical entity occurring in patients of all ages, typically after dog exposure.
While classically considered an infection among immunocompromised patients, most
cases have occurred in previously healthy, immunocompetent persons. Diagnosis
may be rapidly established by PCR, and this test should be considered in
culture-negative cases with associated exposures. Outcome was generally
favorable after a median antibiotic duration of 15 days. Bite infections caused by Capnocytophaga canimorsus are rare. Severe and fatal
infections are more frequently reported in patients with immunodeficiency,
splenectomy or alcohol abuse. We describe the case of a 63-year-old man who
developed flu-like symptoms and presented after some delay with severe sepsis
and purpura fulmis. He was found to be infected with C. canimorsus without a
bite injury and did not demonstrate immunodeficiency or any other typical
predisposition. Despite extensive intensive care, his conditions deteriorated
and he died from multiorgan failure.
LEARNING POINTS: Pet owners with banal, for instance flu-like, symptoms should
urgently seek medical advice when symptoms are unusual.Capnocytophaga canimorsus
infection should be considered and empirical antibiotic therapy immediately
started or adjusted in the presence of purpura fulmis in the absence of
animal bites or immunodeficiency. Capnocytophaga species are gram-negative bacilli that inhabit mammalian oral
surfaces and can cause opportunistic infection, especially in asplenic patients.
The species Capnocytophaga canimorsus is particularly associated with dog bites
and is known to cause endocarditis, meningitis, and sepsis in the general
population. In pregt patients, infections tied to Capnocytophaga species from
human flora have been associated with preterm labor, chorioamnionitis, and
neonatal septicemia. There is little known about the effects of
zoonotically-acquired Capnocytophaga infection in pregt patients. In this
case report, we present a patient with Capnocytophaga bacteremia acquired after
a dog bite associated with profound thrombocytopenia and preterm labor. Dog
bites are common in the United States, and we present basic recommendations for
management of dog bites in pregt patients in order to avoid morbidity
associated with delay in time to antibiotic treatment of infection as described
in this case. Author information:
(1)Department of Emergency and Critical Care Medicine, Kansai Medical University
Medical Center, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8507, Japan. Electronic
address: [email protected].
(2)Department of Emergency and Critical Care Medicine, Kansai Medical University
Medical Center, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8507, Japan. Electronic
address: [email protected].
(3)Department of Emergency and Critical Care Medicine, Kansai Medical University
Medical Center, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8507, Japan. Electronic
address: [email protected].
(4)Department of Emergency and Critical Care Medicine, Kansai Medical University
Medical Center, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8507, Japan. Electronic
address: [email protected].
(5)Department of Emergency and Critical Care Medicine, Kansai Medical University
Medical Center, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8507, Japan. Electronic
address: [email protected].
(6)Department of Plastic and Reconstructive Surgery, Kansai Medical University
Medical Center, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8507, Japan. Electronic
address: [email protected].
(7)Department of Plastic and Reconstructive Surgery, Kansai Medical University
Medical Center, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8507, Japan. Electronic
address: [email protected].
(8)Department of Emergency and Critical Care Medicine, Kansai Medical University
Medical Center, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8507, Japan. Electronic
address: [email protected].
(9)Department of Hematology, Kansai Medical University Medical Center, 10-15
Fumizono-cho, Moriguchi, Osaka 570-8507, Japan. Electronic address:
[email protected].
(10)Department of Emergency and Critical Care Medicine, Kansai Medical
University Hospital, 2-3-1 Shinmachi, Hirakata, Osaka 573-1191, Japan.
Electronic address: [email protected]. |
What is the function of lysozyme? | Lysozymes are an ancient group of antimicrobial enzymes of the innate immune system. Lysozyme activity is a marker of Paneth cell function. | The ICChI is a 35-kDa, glycosylated protein isolated from the latex of the weed
Ipomoea carnea. It displays chitinase and lysozyme activity, which could be
important for the defense against pathogenic fungi, insects and bacteria. The
ICChI enzyme was crystallized, and a diffraction data set was collected from a
single crystal to 1.42 Å resolution. The crystals belong to the primitive
tetragonal space group P43212, with unit-cell parameters a = b = 57.9,
c = 172.0 Å, and α = β = γ = 90°. The structure was elucidated by molecular
replacement method using a mixed model of three homologous structures from the
N-terminal sequence of ICChI. The refined model consists of 272 amino acid
residues and has a Rfactor of 18.93% and Rfree of 22.42%. The protein consists
of a single globular domain with a (α/β)8 triosephosphate isomerase barrel fold.
Three of the consensus sites for N-glycosylation viz., Asn45, Asn172, and Asn194
containing carbohydrate moieties N-Acetylglucosamine (NAG), mannose, fucose, and
xylose. The putative catalytic residues are Asp125, Glu127, and Tyr184. The
crystal structure may provide fundamental information of GH18 family chitinases. The jejunum is the segment of the small intestine responsible for several
metabolism and biotransformation functions. In this report, we have cultured rat
jejunum explants in vitro and integrated them with hepatocyte cultures. We have
also investigated the changes in jejunum function at different locations since
spatial variations in intestinal functions have been reported previously. We
divided the length of the rat jejunum into three distinct regions of
approximately 9 cm each. We defined the regions as proximal (adjacent to the
duodenum), medial, and distal (adjacent to the ileum). Spatiotemporal variations
in functions were observed between these regions within the jejunum. Alkaline
phosphatase activity (a marker of enterocyte function), decreased twofold
between the proximal and distal regions at 4 hr. Lysozyme activity (a marker of
Paneth cell function) increased from the proximal to the distal jejunum by 40%
at 24 hr. Mucin-covered areas, a marker of goblet cell function, increased by
twofold between the proximal and distal segments of the jejunum at 24 hr. When
hepatocytes were integrated with proximal jejunum explants, statistically higher
urea (~2.4-fold) and mucin (57%) production were observed in the jejunum
explants. The integrated intestine-liver cultures can be used as a platform for
future investigations. |
Explain amniotic band syndrome. | Amniotic band syndrome (ABS) is a rare congenital disease with variable manifestations ranging from simple constriction rings at the extremities to major defects such as exencephaly. | The amniotic band syndrome (ABS) refers to the infrequent occurrence of
congenital deformities presumably due to fetal entanglement in strands of
ruptured amniotic sac. The most commonly associated anomalies include
amputations, constriction bands, syndactyly, craniofacial defects, club feet,
and cleft lip. We present a typical case and short literature review of ABS. The
infant reported also had a connective tissue nevus and a cutaneous striated
muscle hamartoma. The amniotic band syndrome is a collection of fetal malformations associated
with fibrous bands that appear to entangle or entrap various fetal parts in
utero, leading to deformation, malformation, or disruption. This syndrome is
given many names yet follows a clearly defined clinical pattern. Misdiagnosis
and inappropriate family counseling are chronic features. This article reviews
the clinical features of the amniotic band syndrome, its epidemiology, and the
status of prenatal and neonatal diagnosis. The spectrum of malformations
associated with amniotic bands is summarized and illustrated. Major etiologic
theories are examined. It is recommended that the clinician involved in the
delivery of any infant manifesting elements of this unusual combination of
defects seek specialized consultation in the pursuit of an accurate and precise
diagnosis and appropriate genetic counseling. The amniotic band syndrome is a collection of congenital deformities presumably
due to rupture of amniotic sac. It appears to cause fetal injury through three
basic mechanisms including malformation, disruption, and deformation. The
associated anomalies vary from minor digital defect to major craniofacial and
visceral defects. They can be categorized as neural tube-like defects,
craniofacial anomalies, limb anomalies, abdominal and thoracic wall defects,
visceral anomalies, and constriction bands. We present two autopsy cases and
discuss the diagnostic features. Our findings support Torpin's theory that the
fibrous constriction bands generated from early rupture of the amnion. An
accurate diagnosis may be achieved by looking for the major features of amniotic
band syndrome and a routine chromosome study and placental examination in cases
with multiple congenital deformities. Amniotic band syndrome is a sporadic condition that occurs in approximately
1:1200 to 1:15,000 live births and that may result in amputations, constrictions
and other deformities of the fetus. Although some cases present with congenital
anomalies that are beyond surgical repair, a selected group of fetuses may show
isolated limb constriction. It has been speculated that, without treatment,
amputation or severe dysfunction of the limb may occur. Despite these potential
complications, surgical treatment for this selected group of fetuses has not
been previously performed. We report two cases that were successfully treated
using novel minimally invasive surgical techniques. The cases involved fetuses
with amniotic band syndrome with associated limb constriction in which the
amniotic band was surgically interrupted to avoid spontaneous amputation of the
extremity. Adequate blood flow distal to the obstruction was preserved and
significant functional improvement of the extremity occurred in both cases,
preserving the limbs. These cases represent the first prenatal surgical
intervention successfully used to treat constricting amniotic bands in humans.
In addition, these cases represent the first time that a non-lethal fetal entity
has been surgically treated in utero. The results of this innovative therapy
will encourage the efforts to continue developing minimally invasive techniques
for the correction of birth defects. BACKGROUND: The amniotic band syndrome is a collection of fetal malformations
associated with fibrous bands that appear to entrap or entangle various fetal
parts in utero and can affect any organ or system and cause a single or multiple
anomalies. The anomaly, acrania, is characterized by partial or complete absence
of the calvarium with abnormal brain tissue development. Literature reports
association of amniotic band syndrome and acrania postnatally, but not diagnosed
prenatally by ultrasound.
CASE: A young woman, gravida 1, para 0, presented for an initial prenatal visit
at 35 weeks' gestation and had a first ultrasound that showed a single
intrauterine pregcy at 36 weeks' gestation. This ultrasound also showed
polyhydramnios, absence of or a very small cerebrum with either anencephaly or
acrania. A targeted ultrasound scan was performed on the following day, which
confirmed acrania in view of the fact that we did see an absence of the flat
bones of the skull with a substantial amount of abnormal brain tissue present
surrounded by a fetal membrane. The patient was counseled, and labor induction
was scheduled with a male infant delivered weighing 1763 g after a spontaneous
vaginal delivery. The infant was diagnosed with acrania, given supportive care,
and died 11 hours later.
CONCLUSION: Diagnosis of cranial bone defects can be established by ultrasound
in the first trimester of pregcy. The prenatal diagnosis of acrania
associated with amniotic bands by transvaginal ultrasound was visualized in the
third trimester in this case; therefore, appropriate counseling and treatment
options were offered. Amniotic band syndrome is an uncommon congenital pathological condition that may
lead to malformations and foetal-infant death. We report an autoptic case. The
patient was a male preterm infant. At 14 weeks of gestation, a routine
ultrasonography showed severe craniofacial anomalies and a close contiguity of
the foetal head with the amnios. The neonate survived three days, after which an
autopsy was carried out. The infant had a frontoparietal meningoencephalocele; a
fibrous band was attached to the skin, close to the meningoencephalocele base.
Cleft lip and palate, nose deformation and agenesis of the right eye were also
present. At the opening of the cranial cavity, occipital hyperostosis was
observed. The herniated brain showed anatomical abnormalities that made
identification of normal structures difficult. Microscopically, the nervous
parenchyma had architectural disorganization and immaturity, and the fibrous
band consisted of amniotic membranes. As evident from this case report, amniotic
band syndrome may cause severe malformations and foetal-infant death. Ethmocephaly is the rarest form of holoprosencephaly, which occurs due to an
incomplete cleavage of the forebrain. Clinically, the disease presents with a
proboscis, hypotelorism, microphthalmos and malformed ears. Amniotic band
syndrome is another rare congenital malformation with ring-like constriction
bands in the limbs, head, face or trunk. We present a case of ethmocephaly with
amniotic band syndrome, which is likely the first of its kind, published in the
literature. Amniotic band syndrome (ABS) is a fetal congenital malformation, affecting
mainly the limbs, but also the craniofacial area and internal organs. Two mains
pathogenic mechanisms are proposed in its genesis. Firstly the early amnion
rupture (exogenous theory) leading to fibrous bands, which wrap up the fetal
body; secondly, the endogenous theory privileges vascular origin, mesoblastic
strings not being a causal agent. The authors believe that the second theory
explain the occurrence of ABS. The outcome of the disease during pregcy
depends on the gravity of the malformations. Interruption of the pregcy is
usually proposed when diagnosis of severe craniofacial and visceral
abnormalities is confirmed. Whereas minor limb defects can be repaired with
postnatal surgery. In case of an isolated amniotic band with a constricted limb,
in utero lysis of the band can be considered to avoid a natural amputation. In
an African country, such treatment is not possible as far as the antenatal
diagnosis. Amniotic band syndrome is a group of sporadic congenital anomalies that involve
the limbs, craniofacial regions and trunk, ranging from simple digital band
constriction to complex craniofacial and central nervous system abnormalities.
Placento-cranial adhesions in amniotic band syndrome are extremely rare, and
severe conditions are associated with high morbidity and mortality rates. In
this study, we pooled placento-cranial adhesion case reports that were published
in the medical literature and added an unpublished case from our institution.
The purpose of this article was to review and discuss the clinical features and
outcomes of placento-cranial adhesions in amniotic band syndrome. Amniotic band syndrome is a rare congenital disorder caused by entrapment of
fetal parts (usually a limb or digits) in fibrous amniotic bands while in utero
that presents with complex multisystem anomalies. The authors report 2 children
with amniotic band syndrome who presented to the ophthalmic unit of the authors'
pediatric hospital. One of them presented with telecanthus, syndactyly,
amputated toes, and unilateral epiphora diagnosed as congenital nasolacrimal
duct obstruction. She was managed conservatively with lacrimal sac massage and
provided with refractive correction while she simultaneously underwent multiple
surgeries for correction of clubfoot and craniosynostosis. The second patient
presented with cleft lip, cleft palate, multiple constriction bands in upper
limbs and fingers with unilateral microphthalmos, microcornea, typical iris
coloboma, and retinochoroidal coloboma, very similar to a case reported in
literature. These 2 cases provide an overview of the clinical spectrum of
ophthalmic manifestations along with their staged optimum rehabilitation. INTRODUCTION: Amniotic band syndrome is a rare congenital disorder with clinical
presentation of constricting bands in different parts of extremities or whole
extremities. Conservative or surgical treatment is provided depending on the
type and severity of the anomaly.
CASE OUTLINE: The paper presents the case of a neonate patient with constriction
bands localized on the left leg. During the second week of life, a surgery was
indicated, and a single-stage multiple Z-plasty was performed to correct the
anomalies on the left lower leg. Postoperative edema in the distal part of the
lower leg was easily managed by incisions and drainage. Two months later, the
correction of the stricture of the left thigh was managed using the same
procedure. The postoperative course was uneventful and the outcome was
satisfactory after a two-year follow-up.
CONCLUSION: Evaluation of a patient with amniotic band syndrome, as well as
diagnosis, monitoring, treatment and postoperative care, should always be
multidisciplinary. A single-stage correction approach provided satisfactory both
functional and aesthetic results. Given many morphological variations of the
syndrome, a decision on the strategy of treatment should be made individually
for each patient. Amniotic band syndrome (ABS) is a rare congenital disease with variable
manifestations ranging from simple constriction rings at the extremities to
major defects such as exencephaly. Here we report the case of a female baby born
full term (39 weeks) from a 35-year-old primiparous mother by cesarean section.
In addition to the constriction rings at the extremities (fingers), the newborn
presented facial malformations and a cranial anomaly suggestive of exencephaly.
Supportive treatment was chosen because of the poor prognosis, and the child
died 5 months later. Depending on the anomaly associated with ABS and its
complexity, as in our case, genetic studies should be performed whenever
possible, and the parents should be informed about the possibility of
recurrences and incompatibility with life. Amniotic band sequence (ABS) is an uncommon and heterogeneous congenital
disorder caused by entrapment of fetal parts by fibrous amniotic bands, causing
distinctive structural abnormalities involving limbs, trunk, and craniofacial
regions. The incidence ranges between 1/1200 and 1/15,000 live births, but is
higher in stillbirths and previable fetuses. The intrinsic theory attributes the
constriction band syndrome as an inherent development defect of embryogenesis
while the extrinsic theory proposes that an early amnion rupture is responsible
for the adherent bands. It is also suggested that amputations and constriction
rings might be due to vascular disturbances. Anomalies resulting from amniotic
bands are quite variable and sometimes may simulate chromosomal abnormalities.
The authors report a case of a 36-week-gestation male neonate who lived for 29
hours after a vaginal delivery with an Apgar score of 8/9/9. The mother was
primipara, and the prenatal was uneventful except for two episodes of urinary
tract infections. The newborn examination depicted multiple anomalies
characterized by exencephaly, bilateral labial cleft with distorted nostrils and
palate cleft. There was also facial skin tag band, exophthalmos with hypoplasia
of the eyelids. The limbs showed distal amputation of the fingers in both hands
and feet, oligodactyly associated with syndactyly in the left foot, ring
constriction in the right leg, the presence of right hyperextension, and
clubfoot. The upper limbs showed length discrepancies. Karyotype analysis was
normal at 46 XY. The authors conclude that the recognition of the malformations
secondary to ABS is important in genetic counseling to prevent misdiagnosis
between chromosomal and secondary disruption disorders. BACKGROUND: Postprocedural amniotic band disruption sequence is a condition that
is associated with intrauterine interventions, and it is characterized by a
constriction of the limbs or umbilical cord by fibrous strands, leading to
edema, amputation, and/or fetal demise.
OBJECTIVE: To evaluate the prevalence of, risk factors for, and the outcome of
postprocedural amniotic band disruption sequence after fetoscopic laser surgery
in twin-twin transfusion syndrome cases.
STUDY DESIGN: All consecutive cases of twin-twin transfusion syndrome treated
with fetoscopic laser coagulation of the vascular anastomoses at our center
between January 2002 and March 2019 were included in the study. The occurrence
of postprocedural amniotic band disruption sequence in these cases was recorded,
and the potential risk factors were analyzed.
RESULTS: Postprocedural amniotic band disruption sequence was detected, at
birth, in 2.2% (15/672) of twin-twin transfusion syndrome cases treated with
fetoscopic laser surgery, in both the recipients (10/15, 67%) and the donors
(5/15, 33%). Postprocedural amniotic band disruption sequence primarily affected
the lower extremities (11/15, 73%) and, less frequently, the upper extremities
(2/15, 13%), both the upper and lower extremities (1/15, 7%), or the umbilical
cord (1/15, 7%). Postprocedural amniotic band disruption sequence led to the
amputation of toes in 5 of 15 cases (33%) and resulted in fetal demise because
of constriction of the umbilical cord in 1 case (7%). The independent risk
factors identified for postprocedural amniotic band disruption sequence were
lower gestational age at laser surgery (odds ratio per week, 1.43; 95%
confidence interval, 1.12-1.79; P=.003) and the presence of postprocedural
chorioamniotic membrane separation on antenatal ultrasound examination (odds
ratio, 41.66; 95% confidence interval, 5.44-319.25; P<.001).
CONCLUSION: The prevalence of postprocedural amniotic band disruption sequence
is low, but, when present, it may lead to severe consequences, with amputation
of extremities or fetal demise occurring in more than one-third of the cases.
Lower gestational age at the time of laser therapy and chorioamniotic membrane
separation are independent risk factors for the postprocedural amniotic band
disruption sequence. |
Which are the main advantages of kallisto against similar methodologies? | Kallisto is a pseudo-alignment algorithm, which is a way of quantifying RNA-sequencing. It's used in RNA-seq because it's much faster and more efficient than other methodologies. | Author information:
(1)Innovative Genomics Initiative, University of California, Berkeley,
California, USA.
(2)Department of Computer Science, University of California, Berkeley,
California, USA.
(3)Faculty of Industrial Engineering, Mechanical Engineering and Computer
Science, University of Iceland, Reykjavik, Iceland.
(4)Department of Mathematics, University of California, Berkeley, California,
USA.
(5)Department of Molecular &Cell Biology, University of California, Berkeley,
California, USA. RNA-Seq technology has been gradually becoming a routine approach for
characterizing the properties of transcriptome in terms of organisms, cell types
and conditions and consequently a big burden has been put on the facet of data
analysis, which calls for an easy-to-learn workflow to cope with the increased
demands from a large number of laboratories across the world. We report a
one-in-all solution called hppRNA, composed of four scenarios such as
pre-mapping, core-workflow, post-mapping and sequence variation detection,
written by a series of individual Perl and R scripts, counting on
well-established and preinstalled software, irrespective of single-end or
paired-end, unstranded or stranded sequencing method. It features six
independent core-workflows comprising the state-of-the-art technology with
dozens of popular cutting-edge tools such as Tophat-Cufflink-Cuffdiff,
Subread-featureCounts-DESeq2, STAR-RSEM-EBSeq, Bowtie-eXpress-edgeR,
kallisto-sleuth, HISAT-StringTie-Ballgown, and embeds itself in Snakemake, which
is a modern pipeline management system. The core function of this pipeline is
turning the raw fastq files into gene/isoform expression matrix and
differentially expressed genes or isoforms as well as the identification of
fusion genes, single nucleotide polymorphisms, long noncoding RNAs and circular
RNAs. Last but not least, this pipeline is specifically designed for performing
the systematic analysis on a huge set of samples in one go, ideally for the
researchers who intend to deploy the pipeline on their local servers. The
scripts as well as the user manual are freely available at
https://sourceforge.net/projects/hpprna/. BACKGROUND: Deconvolution is a mathematical process of resolving an observed
function into its constituent elements. In the field of biomedical research,
deconvolution analysis is applied to obtain single cell-type or tissue specific
signatures from a mixed signal and most of them follow the linearity assumption.
Although recent development of next generation sequencing technology suggests
RNA-seq as a fast and accurate method for obtaining transcriptomic profiles, few
studies have been conducted to investigate best RNA-seq quantification methods
that yield the optimum linear space for deconvolution analysis.
RESULTS: Using a benchmark RNA-seq dataset, we investigated the linearity of
abundance estimated from seven most popular RNA-seq quantification methods both
at the gene and isoform levels. Linearity is evaluated through parameter
estimation, concordance analysis and residual analysis based on a multiple
linear regression model. Results show that count data gives poor parameter
estimations, large intercepts and high inter-sample variability; while TPM value
from Kallisto and Salmon shows high linearity in all analyses.
CONCLUSIONS: Salmon and Kallisto TPM data gives the best fit to the linear model
studied. This suggests that TPM values estimated from Salmon and Kallisto are
the ideal RNA-seq measurements for deconvolution studies. RNA-sequencing has become the gold standard for whole-transcriptome gene
expression quantification. Multiple algorithms have been developed to derive
gene counts from sequencing reads. While a number of benchmarking studies have
been conducted, the question remains how individual methods perform at
accurately quantifying gene expression levels from RNA-sequencing reads. We
performed an independent benchmarking study using RNA-sequencing data from the
well established MAQCA and MAQCB reference samples. RNA-sequencing reads were
processed using five workflows (Tophat-HTSeq, Tophat-Cufflinks, STAR-HTSeq,
Kallisto and Salmon) and resulting gene expression measurements were compared to
expression data generated by wet-lab validated qPCR assays for all protein
coding genes. All methods showed high gene expression correlations with qPCR
data. When comparing gene expression fold changes between MAQCA and MAQCB
samples, about 85% of the genes showed consistent results between RNA-sequencing
and qPCR data. Of note, each method revealed a small but specific gene set with
inconsistent expression measurements. A significant proportion of these
method-specific inconsistent genes were reproducibly identified in independent
datasets. These genes were typically smaller, had fewer exons, and were lower
expressed compared to genes with consistent expression measurements. We propose
that careful validation is warranted when evaluating RNA-seq based expression
profiles for this specific gene set. The recently introduced Kallisto pseudoaligner has radically simplified the
quantification of transcripts in RNA-sequencing experiments. We offer
cloud-scale RNAseq pipelines Arkas-Quantification, and Arkas-Analysis available
within Illumina's BaseSpace cloud application platform which expedites Kallisto
preparatory routines, reliably calculates differential expression, and performs
gene-set enrichment of REACTOME pathways . Due to inherit inefficiencies of
scale, Illumina's BaseSpace computing platform offers a massively parallel
distributive environment improving data management services and data importing.
Arkas-Quantification deploys Kallisto for parallel cloud computations and is
conveniently integrated downstream from the BaseSpace Sequence Read Archive
(SRA) import/conversion application titled SRA Import. Arkas-Analysis annotates
the Kallisto results by extracting structured information directly from source
FASTA files with per-contig metadata, calculates the differential expression and
gene-set enrichment analysis on both coding genes and transcripts. The Arkas
cloud pipeline supports ENSEMBL transcriptomes and can be used downstream from
the SRA Import facilitating raw sequencing importing, SRA FASTQ conversion, RNA
quantification and analysis steps. RNA-seq is a vital method for understanding gene structure and expression
patterns. Typical RNA-seq analysis protocols use sequencing reads of length 50
to 150 nucleotides for alignment to the reference genome and assembly of
transcripts. The resultant transcripts are quantified and used for differential
expression and visualization. Existing tools and protocols for RNA-seq are vast
and diverse; given their differences in performance, it is critical to select an
analysis protocol that is scalable, accurate, and easy to use. Tuxedo, a popular
alignment-based protocol for RNA-seq analysis, has been updated with HISAT2,
StringTie, StringTie-merge, and Ballgown, and the updated protocol outperforms
its predecessor. Similarly, new pseudo-alignment-based protocols like Kallisto
and Sleuth reduce runtime and improve performance. However, these tools are
challenging for researchers lacking command-line experience. Here, we describe
two new RNA-seq analysis protocols, in which all tools are deployed on CyVerse
Cyberinfrastructure with user-friendly graphical user interfaces, and validate
their performance using plant RNA-seq data. © 2018 by John Wiley & Sons, Inc. Rainbow smelt, Osmerus mordax, have an impressive ability to acclimate to very
cold water. Rainbow smelt exposed to cold (<5 °C) for an extended period of time
have faster sustained swimming speeds and increased contraction kinetics in
their myotomal muscle compared to warm acclimated fish. We used RNA Sequencing
reactions (RNA-Seq) to explore how gene expression underlies thermal acclimation
by muscle in these fish. Transcriptome analysis is limited in species that lack
an annotated genome, such as rainbow smelt. The Trinity software package permits
the de novo assembly of a rainbow smelt transcriptome with a modest learning
curve. The transcriptome was then analyzed with Kallisto to quantify the
abundance of each transcript represented in the full transcriptome and Sleuth to
analyze the resulting RNA-seq datasets. Subsequently qPCR was used to explore
patterns of thermal acclimation and gene expression for genes of metabolic and
muscle contractile function. These methodologies revealed shifts in both muscle
and metabolic gene expression that contribute to the thermal acclimation
response in rainbow smelt. In fast-twitch, anaerobic white muscle, slow isoforms
of myosin heavy and light chain tended to be down-regulated with exposure to
cold in myotomal muscle, while fast isoforms were unchanged. Genes associated
with protein turnover and aerobic metabolism were up-regulated in the white
muscle, while those associated with anaerobic metabolism and the cell cycle were
down-regulated. Collectively the results suggest that thermal acclimation to
cold is complex process of apparent shifts in gene expression. |
What methodology does the HercepTest use? | The HercepTest is immunohistochemistry based. | BACKGROUND: HER2 status is a predictive biomarker of response to trastuzumab in
advanced gastric or gastroesophageal junction (GEJ) adenocarcinoma. However,
there is relatively little known about the role of HER2 in resected gastric or
GEJ adenocarcinoma in the Western population.
METHODS: Retrospective, observational, single centre study of patients with
gastric or GEJ adenocarcinoma undergoing surgery with curative intent between
January 2007 and June 2014 in the University Hospital Complex of Santiago de
Compostela. The expression of HER2 was determined by immunohistochemistry (IHC)
using DAKO-HercepTest™ and gene amplification with DuoCISH using a DAKO-DuoCISH
kit. The study of HER2 expression and amplification was carried out in all the
patients and it was correlated with classic clinicopathological parameters,
survival and recurrence pattern.
RESULTS: 106 patients were included. HER2 expression was as follows: 71.7% HER2
negative, 21.7% HER2 equivocal and 6.6% HER2 positive, or with HER2
overexpression. 13.2% of patients (14/106) had HER2 amplification by DuoCISH. A
significant association was seen between overexpression and amplification of
HER2 (p < 0.001).HER2 positivity was associated with the intestinal subtype (p =
0.010) and a low grade of differentiation (p = 0.018). Likewise, HER2 was
significantly associated with a worse prognosis: overall survival (OS) 32.3
months HER2 positive versus 93.9 months HER2 negative (HR 0.42; confidence
interval 95% 0.18-0.93; p = 0.028); and the presence of distant metastasis
without accompanying locoregional recurrence (p = 0.048).
CONCLUSION: HER2 status defines a subgroup with differentiated
clinicopathological characteristics, worse prognosis and distant dissemination,
without accompanying locoregional recurrence, in patients with resected gastric
or GEJ adenocarcinoma operated on in a Western population. |
Which R/Bioconductor package has been developed for gene expression signature searching? | SignatureSearch is an R/Bioconductor package that integrates a suite of existing and novel algorithms into an analysis environment for gene expression signature (ESE) searching combined with functional enrichment analysis (FEA) and visualization methods to facilitate the interpretation of the search results. | signatureSearch is an R/Bioconductor package that integrates a suite of existing
and novel algorithms into an analysis environment for gene expression signature
(GES) searching combined with functional enrichment analysis (FEA) and
visualization methods to facilitate the interpretation of the search results. In
a typical GES search (GESS), a query GES is searched against a database of GESs
obtained from large numbers of measurements, such as different genetic
backgrounds, disease states and drug perturbations. Database matches sharing
correlated signatures with the query indicate related cellular responses
frequently governed by connected mechanisms, such as drugs mimicking the
expression responses of a disease. To identify which processes are predomitly
modulated in the GESS results, we developed specialized FEA methods combined
with drug-target network visualization tools. The provided analysis tools are
useful for studying the effects of genetic, chemical and environmental
perturbations on biological systems, as well as searching single cell GES
databases to identify novel network connections or cell types. The
signatureSearch software is unique in that it provides access to an integrated
environment for GESS/FEA routines that includes several novel search and
enrichment methods, efficient data structures, and access to pre-built GES
databases, and allowing users to work with custom databases. |
Which molecule is targeted by Upadacitinib? | Upadacitinib is a Janus kinase 1 inhibitor developed for treatment of moderate to severe rheumatoid arthritis. | AIMS: Upadacitinib (ABT-494) is a selective Janus kinase 1 inhibitor being
developed for treatment of auto-immune inflammatory disorders. This work
evaluated effects of high-fat meal, cytochrome P450 (CYP) 3A inhibition, CYP
induction, and organic anion transporting polypeptide (OATP) 1B inhibition on
upadacitinib pharmacokinetics.
METHODS: Two Phase 1 evaluations were conducted, each in 12 healthy subjects. In
Study 1, using a randomized, two-sequence crossover design, a 3 mg dose of
upadacitinib (immediate-release capsules) was administered alone under fasting
conditions, after high-fat meal, or on Day 4 of a 6-day regimen of 400 mg
once-daily ketoconazole. In Study 2, a 12 mg upadacitinib dose was administered
alone, with the first, and with the eighth dose of a 9-day regimen of rifampin
600 mg once daily. Upadacitinib plasma concentrations were characterized.
RESULTS: Administration of upadacitinib immediate-release capsules after a
high-fat meal decreased upadacitinib Cmax by 23% and had no impact on
upadacitinib AUC relative to the fasting conditions. Ketoconazole (strong CYP3A
inhibitor) increased upadacitinib Cmax and AUC by 70% and 75%, respectively.
Multiple doses of rifampin (broad CYP inducer) decreased upadacitinib Cmax and
AUC by approximately 50% and 60%, respectively. A single dose of rifampin (also
an OATP1B inhibitor) had no effect on upadacitinib AUC. Upadacitinib was well
tolerated when co-administered with ketoconazole, rifampin, or after a high-fat
meal.
CONCLUSIONS: Strong CYP3A inhibition and broad CYP induction result in a weak
and moderate effect, respectively, on upadacitinib exposures. OATP1B inhibition
and administration of upadacitinib immediate-release formulation with food does
not impact upadacitinib exposure. Exposure-response analyses of QT data from early-stage clinical studies
represent a valuable tool to assess the QT prolongation potential for drugs in
development in lieu of standalone thorough QT (TQT) studies. However,
demonstrating adequate electrocardiogram assay sensitivity can be challenging in
the absence of a positive pharmacological control. Upadacitinib is a Janus
kinase 1 inhibitor currently being evaluated in phase III rheumatoid arthritis
trials. Exposure-response analyses to evaluate the QT prolongation potential for
upadacitinib from phase I trials and the utility of the effect of food on QTcF
to demonstrate ECG assay sensitivity are presented. The analyses demonstrated no
effect of upadacitinib on QT interval and confirmed the sensitivity of the ECG
assay to detect the small QT shortening effect caused by food. Lack of bias from
manual ECG adjudication was also demonstrated. These analyses supported
requesting a waiver for the regulatory requirement for a dedicated thorough QT
study for upadacitinib. The past three decades have witnessed remarkable advances in our ability to
target specific elements of the immune and inflammatory response, fuelled by
advances in both biotechnology and disease knowledge. As well as providing
superior treatments for immune-mediated inflammatory diseases (IMIDs), such
therapies also offer unrivalled opportunities to study the underlying
immunopathological basis of these conditions.In this review, we explore recent
approaches to the treatment of IMIDs and the insights to pathobiology that they
provide. We review novel biologic agents targeting the T-helper 17 axis,
including therapies directed towards interleukin (IL)-17 (secukinumab,
ixekizumab, bimekizumab), IL-17R (brodalumab), IL-12/23p40 (ustekinumab,
briakinumab) and IL-23p19 (guselkumab, tildrakizumab, brazikumab, risankizumab,
mirikizumab). We also present an overview of biologics active against type I and
II interferons, including sifalumumab, rontalizumab, anifrolumab and
fontolizumab. Emerging strategies to interfere with cellular adhesion processes
involved in lymphocyte recruitment are discussed, including both integrin
blockade (natalizumab, vedolizumab, etrolizumab) and sphingosine-1-phosphate
receptor inhibition (fingolimod, ozanimod). We summarise the development and
recent application of Janus kinase (JAK) inhibitors in the treatment of IMIDs,
including first-generation pan-JAK inhibitors (tofacitinib, baricitinib,
ruxolitinib, peficitinib) and second-generation selective JAK inhibitors
(decernotinib, filgotinib, upadacitinib). New biologics targeting B-cells
(including ocrelizumab, veltuzumab, tabalumab and atacicept) and the development
of novel strategies for regulatory T-cell modulation (including low-dose IL-2
therapy and Tregitopes) are also discussed. Finally, we explore recent
biotechnological advances such as the development of bispecific antibodies
(ABT-122, COVA322), and their application to the treatment of IMIDs. BACKGROUND: There is a great unmet clinical need for efficacious, tolerable,
economical and orally administrated drugs for the treatment of inflammatory
bowel disease (IBD). New therapeutic avenues have become possible including the
development of medications that target specific genetic pathways found to be
relevant in other immune mediated diseases.
AIMS: To provide an overview of recent clinical trials for new generation oral
targeted medications that may have a future role in IBD management.
METHODS: Pubmed and Medline searches were performed up to 1 March 2018 using
keywords: "IBD", "UC", "CD", "inflammatory bowel disease" "ulcerative colitis",
"Crohn's disease" in combination with "phase", "study", "trial" and "oral". A
manual search of the clinical trial register, article reference lists, abstracts
from meetings of Digestive Disease Week, United European Gastroenterology Week
and ECCO congress were also conducted.
RESULTS: In randomised controlled trials primary efficacy endpoints were met for
tofacitinib (JAK 1/3 inhibitor-phase III), upadacitinib (JAK 1 inhibitor-phase
II) and AJM300 (α4-integrin antagonist-phase II) in ulcerative colitis. Ozanimod
(S1P receptor agonist-phase II) also demonstrated clinical remission. For
Crohn's disease, filgotinib (JAK1 inhibitor-phase II) met primary endpoints and
laquinimod (quinolone-3-carboxide small molecule-phase II) was also efficacious.
Trials using mongersen (SMAD7 inhibitor) and vidofludimus (dihydroorotate
dehydrogenase inhibitor) have been halted.
CONCLUSIONS: This is potentially the start of an exciting new era in which
multiple therapeutic options are at the disposal of physicians to treat IBD on
an individualised basis. Head-to-head studies with existing treatments and
longer term safety data are needed for this to be possible. Upadacitinib is a Janus kinase 1 inhibitor under development for the treatment
of several inflammatory disorders including rheumatoid arthritis (RA).
Upadacitinib was administered in the phase 2 RA trials primarily as twice-daily
regimens of an immediate-release (IR) formulation. The upadacitinib
extended-release (ER) formulation was developed to enable once-daily dosing. In
the present study, upadacitinib pharmacokinetics were characterized after the
administration of single and multiple once-daily doses of the ER formulation in
healthy subjects relative to single and multiple twice-daily doses of the IR
formulation. Increase in upadacitinib exposure was dose-proportional over the
evaluated 15- to 30-mg ER dose range. Single 15- and 30-mg ER doses provided
equivalent AUC0-inf compared with single 12- and 24-mg IR doses, respectively. A
high-fat breakfast increased upadacitinib ER Cmax and AUC0-inf by only 20% and
17%, respectively, relative to fasting conditions. The median time to peak
plasma concentrations was 2 to 4 hours for the ER formulation, and steady state
was achieved by day 4 of once-daily dosing. Doses of 15 and 30 mg once daily
using the ER formulation provided equivalent AUC0-24 , comparable Cmax and Cmin
, and a fluctuation index over a 24-hour period at steady state similar to 6 and
12 mg twice daily, respectively, using the IR formulation. These results
supported the use of upadacitinib 15- and 30-mg doses of the ER formulation in
the phase 3 trials in RA. BACKGROUND: Upadacitinib is a selective inhibitor of Janus kinase 1 and was
efficacious in phase 2 studies in patients with moderate-to-severe rheumatoid
arthritis. We aimed to assess the efficacy of upadacitinib in patients with
inadequate response to conventional synthetic disease-modifying anti-rheumatic
drugs (csDMARDs).
METHODS: This study is a double-blind, placebo-controlled trial at 150 sites in
35 countries. We enrolled patients aged 18 years or older with active rheumatoid
arthritis for 3 months or longer, who had received csDMARDs for at least 3
months with a stable dose for at least 4 weeks before study entry, and had an
inadequate response to at least one of the following csDMARDs: methotrexate,
sulfasalazine, or leflunomide. Using interactive response technology, we
randomly assigned patients receiving stable background csDMARDs (2:2:1:1) to
receive a once-daily extended-release formulation of upadacitinib 15 mg or 30
mg, or placebo, for 12 weeks. Patients, investigators, and the funder were
masked to allocation. After 12 weeks, patients taking placebo received 15 mg or
30 mg of upadacitinib once daily, according to the prespecified randomisation
assignment. The primary endpoints were the proportion of patients at week 12 who
achieved 20% improvement in American College of Rheumatology criteria (ACR20),
and a 28-joint disease activity score using C-reactive protein (DAS28[CRP]) of
3·2 or less. We did efficacy analyses in the full analysis set of all randomly
assigned patients who received at least one dose of study drug, and used
non-responder imputation for assessment of the primary outcomes. This study is
registered with ClinicalTrials.gov, number NCT02675426.
FINDINGS: Between Dec 17, 2015, and Dec 22, 2016, 1083 patients were assessed
for eligibility, of whom 661 were recruited and randomly assigned to receive
upadacitinib 15 mg (n=221), upadacitinib 30 mg (n=219), or placebo (n=221). All
patients received at least one dose of study drug, and 618 (93%) completed 12
weeks of treatment. At week 12, ACR20 was achieved by 141 (64%; 95% CI 58-70) of
221 patients receiving upadacitinib 15 mg and 145 (66%; 60-73) of 219 patients
receiving upadacitinib 30 mg, compared with 79 (36%; 29-42) of 221 patients
receiving placebo (p<0·0001 for each dose vs placebo). DAS28(CRP) of 3·2 or less
was met by 107 (48%; 95% CI 42-55) patients receiving upadacitinib 15 mg and 105
(48%; 41-55) patients receiving upadacitinib 30 mg, compared with 38 (17%;
12-22) patients receiving placebo (p<0·0001 for each dose vs placebo). Adverse
events were reported in 125 (57%) of 221 patients receiving upadacitinib 15 mg,
118 (54%) of 219 patients receiving upadacitinib 30 mg, and 108 (49%) of 221
patients receiving placebo. The most frequently reported adverse events (≥5% of
patients in any group) were nausea (16 [7%] of 221 in the upadacitinib 15 mg
group; three [1%] of 219 in the upadacitinib 30 mg group; and seven [3%] of 221
in the placebo group), nasopharyngitis (12 [5%]; 13 [6%]; and nine [4%]), upper
respiratory tract infection (12 [5%]; 12 [5%]; and nine [4%]), and headache
(nine [4%]; seven [3%]; and 12 [5%]). More infections were reported for
upadacitinib (64 [29%] of 221 patients receiving 15 mg and 69 [32%] of 219
patients receiving 30 mg) versus placebo (47 [21%] of 221 patients). There were
three herpes zoster infections (one [<1%] in the placebo group, one [<1%] in the
upadacitinib 15 mg group, and one [<1%] in the upadacitinib 30 mg group) and one
primary varicella zoster virus infection (one [<1%] in the upadacitinib 30 mg
group), two maligcies (both in the upadacitinib 30 mg group), one adjudicated
major adverse cardiovascular event (in the upadacitinib 30 mg group), and five
serious infections (one [<1%] in the placebo group, one [<1%] in the
upadacitinib 15 mg group, three [1%] in the upadacitinib 30 mg group). No deaths
were reported during the trial.
INTERPRETATION: Patients with moderately to severely active rheumatoid arthritis
who received upadacitinib (15 mg or 30 mg) in combination with csDMARDs showed
significant improvements in clinical signs and symptoms.
FUNDING: AbbVie Inc. BACKGROUND: Phase 2 studies with upadacitinib, a selective Janus kinase 1 (JAK1)
inhibitor, have shown safety and efficacy in the treatment of patients with
active rheumatoid arthritis. We did this study to further assess the safety and
efficacy of upadacitinib in patients with an inadequate response to biologic
disease-modifying anti-rheumatic drugs (bDMARDs).
METHODS: We did this double-blind, randomised controlled phase 3 trial at 153
sites in 26 countries. Patients were aged 18 years or older, had active
rheumatoid arthritis and previous inadequate response or intolerance to bDMARDs,
and were receiving concomitant background conventional synthetic DMARDS
(csDMARDs). We randomly assigned patients (2:2:1:1) by interactive response
technology to receive once-daily oral extended-release upadacitinib 15 mg or 30
mg or placebo for 12 weeks, followed by upadacitinib 15 mg or 30 mg from week 12
onwards. The two separate primary endpoints were the proportions of patients
achieving a 20% improvement in American College of Rheumatology criteria (ACR20)
at week 12 and the proportion of patients achieving a 28-joint disease activity
score using C-reactive protein (DAS28[CRP]) of 3·2 or less at week 12. Efficacy
and safety analyses were done in the modified intention-to-treat population of
all patients who received at least one dose of study drug. Data are presented up
to week 24 of this ongoing study. The trial is registered with
ClinicalTrials.gov (NCT02706847).
FINDINGS: Between March 15, 2016, and Jan 10, 2017, 499 patients were randomly
assigned (n=165 upadacitinib 15 mg; n=165 upadacitinib 30 mg; n=85 placebo then
upadacitinib 15 mg; and n=84 placebo then upadacitinib 30 mg) and one patient
was withdrawn from the 15 mg upadacitinib group before the start of study
treatment. Mean disease duration was 13·2 years (SD 9·5); 235 (47%) of 498
patients had received one previous bDMARD, 137 (28%) had received two, and 125
(25%) had received at least three; 451 (91%) patients completed treatment up to
week 12 and 419 (84%) patients completed treatment up to week 24. At week 12,
ACR20 was achieved by 106 (65%; 95% CI 57-72) of 164 patients receiving
upadacitinib 15 mg and 93 (56%; 49-64) of 165 patients receiving upadacitinib 30
mg compared with 48 (28%; 22-35) of 169 patients receiving placebo (p<0·0001 for
each dose vs placebo). DAS28(CRP) of 3·2 or less was achieved by 71 (43%; 95% CI
36-51) of 164 patients receiving upadacitinib 15 mg and 70 (42%; 35-50) of 165
patients receiving upadacitinib 30 mg versus 24 (14%; 9-20) of 169 patients
receiving placebo (p<0·0001 for each dose vs placebo). Up to week 12, overall
numbers of patients with adverse events were similar for the placebo group (95
[56%] of 169) and the upadacitinib 15 mg group (91 [55%] of 164), but higher in
the upadacitinib 30 mg group (111 [67%] of 165). At week 12, the most common
adverse events occurring in at least 5% of patients in any treatment group were
upper respiratory tract infection (13 [8%] of 169 in the placebo group; 13 [8%]
of 164 in the upadacitinib 15 mg group; ten [6%] of 165 in the upadacitinib 30
mg group), nasopharyngitis (11 [7%]; seven [4%]; nine [5%]), urinary tract
infection (ten [6%]; 15 [9%]; nine [5%]), and worsening of rheumatoid arthritis
(ten [6%]; four [2%]; six [4%]). The number of patients with serious adverse
events was higher in the upadacitinib 30 mg group (12 [7%]) than in the
upadacitinib 15 mg group (eight [5%]); no serious adverse events were reported
in patients receiving placebo. More patients in the upadacitinib 30 mg group had
serious infections, herpes zoster, and adverse events leading to discontinuation
than in the upadacitinib 15 mg and placebo groups. During the placebo-controlled
phase of the study, one case of pulmonary embolism, three maligcies, one
major adverse cardiovascular event, and one death were reported in patients
receiving upadacitinib; none were reported in patients receiving placebo.
INTERPRETATION: Both doses of upadacitinib led to rapid and significant
improvements compared with placebo over 12 weeks in patients with refractory
rheumatoid arthritis.
FUNDING: AbbVie Inc. Upadacitinib is a novel selective oral Janus kinase 1 (JAK) inhibitor being
developed for treatment of several inflammatory diseases. Oral contraceptives
are anticipated to be a common concomitant medication in the target patient
populations. This study was designed to evaluate the effect of multiple doses of
upadacitinib on the pharmacokinetics of ethinylestradiol and levonorgestrel in
healthy female subjects. This phase I, single-center, open-label, 2-period
crossover study evaluated the effect of multiple doses of 30 mg once daily
extended-release upadacitinib on the pharmacokinetics of a single oral dose of
ethinylestradiol/levonorgestrel (0.03/0.15 mg; administered alone in period 1
and on day 12 of a 14-day regimen of upadacitinib in period 2) in 22 healthy
female subjects. The ratios (90% confidence intervals) for maximum plasma
concentration and area under the plasma drug concentration-time curve from time
zero to infinity following administration of ethinylestradiol/levonorgestrel
with upadacitinib compared with administration of ethinylestradiol/
levonorgestrel alone were 0.96 (0.89-1.02) and 1.1 (1.04-1.19), respectively,
for ethinylestradiol, and 0.96 (0.87-1.06) and 0.96 (0.85-1.07), respectively,
for levonorgestrel. The harmonic mean terminal half-life for ethinylestradiol
(7.7 vs 7.0 hours) and levonorgestrel (37.1 vs 33.1 hours) was similar in the
presence and absence of upadacitinib. Ethinylestradiol and levonorgestrel were
bioequivalent in the presence and absence of upadacitinib. Therefore,
upadacitinib can be administered concomitantly with oral contraceptives
containing ethinylestradiol or levonorgestrel. OBJECTIVES: We assessed the relative efficacy and safety of once-daily
administration of 15 and 30 mg upadacitinib (a JAK1-selective inhibitor) in
patients with active rheumatoid arthritis (RA).
METHODS: We conducted a Bayesian network meta-analysis to combine the direct and
indirect evidence from randomized controlled trials (RCTs) that examined the
efficacy and safety of upadacitinib in patients with active RA.
RESULTS: Five RCTs involving 4381 patients met the inclusion criteria. There
were 15 pairwise comparisons, including eight direct comparisons and six
interventions. The ACR20 response rate was significantly higher in the
upadacitinib 15 and 30 mg + MTX (methotrexate) groups than in the MTX group (OR:
4.98, 95% CrI: 2.66-10.10; OR: 4.73, 95% CrI: 2.25-10.98). Adalimumab
40 mg + MTX, upadacitinib 30 mg, and upadacitinib 15 mg groups showed
a significantly higher ACR20 response rate than did the MTX group. Ranking
probability based on the surface under the cumulative ranking curve (SUCRA)
indicated that upadacitinib 15 mg + MTX was likely to achieve the best ACR20
response rate (SUCRA = 0.838), followed by upadacitinib 30 mg + MTX, adalimumab
40 mg + MTX, upadacitinib 30 mg, upadacitinib 15 mg, and MTX (SUCRA = 0.784,
0.495, 0.471, 0.404, and 0.008, respectively). The safety based on the number of
serious adverse events (SAEs) did not differ significantly among the six
interventions.
CONCLUSIONS: Upadacitinib 15 and 30 mg administration once daily in combination
with MTX was the most efficacious intervention for active RA, with no
significant risk for SAEs. BACKGROUND: Anti-cytokine therapies such as adalimumab, tocilizumab, and the
small molecule JAK inhibitor tofacitinib have proven that cytokines and their
subsequent downstream signaling processes are important in the pathogenesis of
rheumatoid arthritis. Tofacitinib, a pan-JAK inhibitor, is the first approved
JAK inhibitor for the treatment of RA and has been shown to be effective in
managing disease. However, in phase 2 dose-ranging studies tofacitinib was
associated with dose-limiting tolerability and safety issues such as anemia.
Upadacitinib (ABT-494) is a selective JAK1 inhibitor that was engineered to
address the hypothesis that greater JAK1 selectivity over other JAK family
members will translate into a more favorable benefit:risk profile. Upadacitinib
selectively targets JAK1 dependent disease drivers such as IL-6 and IFNγ, while
reducing effects on reticulocytes and natural killer (NK) cells, which
potentially contributed to the tolerability issues of tofacitinib.
METHODS: Structure-based hypotheses were used to design the JAK1 selective
inhibitor upadacitinib. JAK family selectivity was defined with in vitro assays
including biochemical assessments, engineered cell lines, and cytokine
stimulation. In vivo selectivity was defined by the efficacy of upadacitinib and
tofacitinib in a rat adjuvant induced arthritis model, activity on reticulocyte
deployment, and effect on circulating NK cells. The translation of the
preclinical JAK1 selectivity was assessed in healthy volunteers using ex vivo
stimulation with JAK-dependent cytokines.
RESULTS: Here, we show the structural basis for the JAK1 selectivity of
upadacitinib, along with the in vitro JAK family selectivity profile and
subsequent in vivo physiological consequences. Upadacitinib is ~ 60 fold
selective for JAK1 over JAK2, and > 100 fold selective over JAK3 in cellular
assays. While both upadacitinib and tofacitinib demonstrated efficacy in a rat
model of arthritis, the increased selectivity of upadacitinib for JAK1 resulted
in a reduced effect on reticulocyte deployment and NK cell depletion relative to
efficacy. Ex vivo pharmacodynamic data obtained from Phase I healthy volunteers
confirmed the JAK1 selectivity of upadactinib in a clinical setting.
CONCLUSIONS: The data presented here highlight the JAK1 selectivity of
upadacinitinib and supports its use as an effective therapy for the treatment of
RA with the potential for an improved benefit:risk profile. Upadacitinib is a selective Janus kinase 1 inhibitor being developed for the
treatment of several inflammatory autoimmune diseases, including rheumatoid
arthritis. Upadacitinib is a nonsensitive substrate for metabolism by cytochrome
P450 3A enzymes. This open-label, single-dose, multicenter study assessed the
pharmacokinetics of upadacitinib following oral administration of a single 15-mg
dose of the upadacitinib extended-release formulation in subjects with mild (n =
6) and moderate (n = 6) hepatic impairment relative to demographically matched
healthy subjects (n = 6). Subjects were assigned to 1 of the 3 groups according
to the Child-Pugh classification. Relative to subjects with normal hepatic
function, the ratios (90% confidence intervals) of upadacitinib area under the
plasma concentration-versus-time profile from time 0 to infinity (AUCinf ) for
subjects with mild and moderate hepatic impairment were 1.28 (0.91-1.79) and
1.24 (0.87-1.76), respectively. The central ratios of upadacitinib maximum
observed concentration (Cmax ) were 1.04 (0.77-1.39) and 1.43 (1.05-1.95) in
subjects with mild and moderate hepatic impairment, respectively, compared with
subjects with normal hepatic function. No clinically significant changes in
vital signs or hematology measurements were observed, and no new safety events
were identified in this study. These results indicate that mild and moderate
hepatic impairment has no clinically relevant effect on upadacitinib
pharmacokinetics. BACKGROUND: Upadacitinib, an oral Janus kinase (JAK)1-selective inhibitor,
showed efficacy in combination with stable background conventional synthetic
disease-modifying antirheumatic drugs (csDMARDs) in patients with rheumatoid
arthritis who had an inadequate response to DMARDs. We aimed to evaluate the
safety and efficacy of upadacitinib monotherapy after switching from
methotrexate versus continuing methotrexate in patients with inadequate response
to methotrexate.
METHODS: SELECT-MONOTHERAPY was conducted at 138 sites in 24 countries. The
study enrolled adults (≥18 years) who fulfilled the 2010 American College of
Rheumatology (ACR)-European League Against Rheumatism (EULAR) classification
criteria for rheumatoid arthritis. Patients with active rheumatoid arthritis
despite stable methotrexate were randomly assigned 2:2:1:1 to switch to
once-daily monotherapy of of upadacitinib or to continue methotrexate at their
existing dose as blinded study drug; starting from week 14, patients assigned to
continue methotrexate were switched to 15 mg or 30 mg once-daily upadacitinib
per prespecified random assignment at baseline. The primary endpoints in this
report are proportion of patients achieving 20% improvement in the ACR criteria
(ACR20) at week 14, and proportion achieving low disease activity defined as
28-joint Disease Activity Score using C-reactive protein (DAS28[CRP]) of 3·2 or
lower, both with non-responder imputation at week 14. Outcomes were assessed in
patients who received at least one dose of study drug. This study is active but
not recruiting and is registered with ClinicalTrials.gov, number NCT02706951.
FINDINGS: Patients were screened between Feb 23, 2016, and May 19, 2017 and 648
were randomly assigned to treatment. 598 (92%) completed week 14. At week 14, an
ACR20 response was achieved by 89 (41%) of 216 patients (95% CI 35-48) in the
continued methotrexate group, 147 (68%) of 217 patients (62-74) receiving
upadacitinib 15 mg, and 153 (71%) of 215 patients (65-77) receiving upadacitinib
30 mg (p<0·0001 for both doses vs continued methotrexate). DAS28(CRP) 3·2 or
lower was met by 42 (19%) of 216 (95% CI 14-25) in the continued methotrexate
group, 97 (45%) of 217 (38-51) receiving upadacitinib 15 mg, and 114 (53%) of
215 (46-60) receiving upadacitinib 30 mg (p<0·0001 for both doses vs continued
methotrexate). Adverse events were reported in 102 patients (47%) on continued
methotrexate, 103 (47%) on upadacitinib 15 mg, and 105 (49%) on upadacitinib 30
mg. Herpes zoster was reported by one (<1%) patient on continued methotrexate,
three (1%) on upadacitinib 15 mg, and six (3%) on upadacitinib 30 mg. Three
maligcies (one [<1%] on continued methotrexate, two [1%] on upadacitinib 15
mg), three adjudicated major adverse cardiovascular events (one [<1%] on
upadacitinib 15 mg, two [<1%] on upadacitinib 30 mg), one adjudicated pulmonary
embolism (<1%; upadacitinib 15 mg), and one death (<1%; upadacitinib 15 mg,
haemorrhagic stroke [ruptured aneurysm]) were reported in the study.
INTERPRETATION: Upadacitinib monotherapy showed statistically significant
improvements in clinical and functional outcomes versus continuing methotrexate
in this methotrexate inadequate-responder population. Safety observations were
similar to those in previous upadacitinib rheumatoid arthritis studies.
FUNDING: AbbVie Inc, USA. Orally bioavailable inhibitors of the tyrosine kinases (TYKs), also referred to
as Janus kinases (JAKs), are being evaluated for the treatment of patients with
Crohn's disease (CD), ulcerative colitis (UC), and other chronic inflammatory
disorders. To date, three JAK inhibitors have been tested in patients with
moderate-to-severe CD: tofacitinib (pan-JAK inhibitor), filgotinib (JAK1
inhibitor) and upadacitinib (JAK1 inhibitor). Clinical development of
tofacitinib was discontinued in CD because the primary endpoint of clinical
remission in the phase II induction and maintece trials was not met, although
outcomes may have been influenced by trial design flaws and a high placebo rate
was noted. In contrast, filgotinib did meet its primary endpoint of clinical
remission at week 10 in the phase II FITZROY trial, in addition to several other
clinically important secondary outcomes, spurring a subsequent larger phase III
trial. Following promising results for upadacitinib in its phase II trial,
larger phase III trials were also initiated to corroborate the efficacy results.
Although JAK inhibitors appear to have an acceptable safety profile, higher
rates of infections compared to placebo were noted. Overall, JAK inhibitors
constitute a new promising class of drugs, given the efficacy signals observed
in pivotal clinical trials in several chronic inflammatory diseases. Here we
review the existing evidence on the pharmacology, safety and efficacy of
JAK-STAT inhibitors that are currently under investigation for the treatment of
patients with CD. The aim of this study was to characterize the effects of upadacitinib, a Janus
kinase 1 inhibitor, on in vivo activity of different cytochrome P450 (CYP)
enzymes using a cocktail approach. Healthy subjects (n = 20) received single
oral doses of the modified Cooperstown 5+1 cocktail drugs (midazolam [CYP3A],
caffeine [CYP1A2], warfarin + vitamin K [CYP2C9], omeprazole [CYP2C19], and
dextromethorphan [CYP2D6]) without upadacitinib and on day 11 (midazolam) or 12
(all other probes) of a 15-day regimen of upadacitinib 30 mg once daily
(extended-release formulation). Serial blood samples and 12-hour urine samples
were collected for assays of the probe substrates and select metabolites. The
ratio (90%CI) of area under the plasma concentration-time curve from time 0 to
infinity (AUCinf ) central values when the cocktail drugs were administered with
upadacitinib relative to when administered alone were 0.74 (0.68-0.80) for
midazolam, 1.22 (1.15-1.29) for caffeine, 1.11 (1.07-1.15) for S-warfarin, 1.07
(0.95-1.22) for dextromethorphan, and 0.82 (0.72-0.94) for omeprazole. The ratio
(90%CI) was 1.09 (1.00-1.19) for 5-hydroxy-omeprazole to omeprazole AUCinf ratio
and 1.17 (0.97-1.41) for dextromethorphan to dextrorphan 12-hour molar urinary
ratio. Upadacitinib 30 mg once daily (a dose that is twice the optimal dose in
rheumatoid arthritis based on phase 3 results) has a limited effect on CYP3A
activity (26% decrease in exposure of midazolam, a sensitive CYP3A substrate)
and no relevant effects on CYP1A2, CYP2C9, CYP2C19, or CYP2D6 activity in vivo.
No clinically relevant changes in plasma exposures are expected for drugs that
are substrates for the evaluated CYP enzymes when coadministered with
upadacitinib. The past decade has witnessed an explosion in trial data on JAK inhibitors
(JAKi). These small molecules target the Janus kinase - signal transducer and
activator of transcription (JAK-STAT) pathway, blocking crucial cytokines across
a septum of rheumatic diseases. As a class, JAKi are beginning to demonstrate
efficacy on par, if not superior to biologics. Two first generation JAKi are
licensed for use in inflammatory arthritis; tofacitinib and baricitinib.
Next-generation JAKi have been designed with selective affinity for one JAK
enzymes, the aim to reduce unwanted adverse effects without declining clinical
efficacy. Emerging data with selective JAK1 inhibitors upadacitinib and
filgotinib looks very promising. Despite differences in selectivity between
JAKi, an overlap exists in their safety profiles. Across the class, a
characteristic safety signal is emerging with viral opportunistic infections,
particularly herpes zoster. Post marketing drug surveillance will be essential
in evaluating the long-term risk with these agents. Upadacitinib is a selective Janus Kinase 1 inhibitor which is being developed
for the treatment of several inflammatory diseases including rheumatoid
arthritis. Upadacitinib was evaluated in Phase 3 studies as an oral
extended-release (ER) formulation administered once daily. The purpose of this
study was to develop a level A in vitro-in vivo correlation (IVIVC) for
upadacitinib ER formulation. The pharmacokinetics of four upadacitinib
extended-release formulations with different in vitro release characteristics
and an immediate-release capsule formulation of upadacitinib were evaluated in
20 healthy subjects in a single-dose, randomized, crossover study. In vivo
pharmacokinetic data and in vitro dissolution data (USP Dissolution Apparatus 1;
pH 6.8; 100 rpm) were used to establish a level A IVIVC. Three formulations were
used to establish the IVIVC, and the fourth formulation was used for external
validation. A non-linear IVIVC best described the relationship between
upadacitinib in vitro dissolution and in vivo absorption profiles. The absolute
percent prediction errors (%PE) for upadacitinib Cmax and AUC were less than 10%
for all three formulations used to establish the IVIVC, as well as for the %PE
for the external validation formulation and the overall mean internal
validation. Model was cross-validated using the leave-one-out approach; all
evaluated cross-validation runs met the regulatory acceptance criteria. A level
A IVIVC was successfully developed and validated for upadacitinib ER
formulation, which meets the FDA and EMA regulatory validation criteria and can
be used as surrogate for in vivo bioequivalence. Inhibition of Janus kinases [JAKs] in Crohn's disease [CD] patients has shown
conflicting results in clinical trials. Tofacitinib, a pan-JAK inhibitor, showed
efficacy in ulcerative colitis [UC] and has been approved for the treatment of
patients with moderate to severe UC. In contrast, studies in CD patients were
disappointing and the primary end point of clinical remission could not be met
in the respective phase II induction and maintece trials. Subsequently, the
clinical development of tofacitinib was discontinued in CD. In contrast,
efficacy of filgotinib, a selective JAK1 inhibitor, in CD patients was
demonstrated in the randomized, double-blinded, placebo-controlled phase II
FITZROY study. Upadacitinib also showed promising results in a phase II trial in
moderate to severe CD. Subsequently, phase III programmes in CD have been
initiated for both substances, which are still ongoing. Several newer molecules
of this class of orally administrated immunosuppressants are being tested in
clinical programmes. The concern of side effects of systemic JAK inhibition is
addressed by either exclusively intestinal action or higher selectivity [Tyk2
inhibitors]. In general, JAK inhibitors constitute a new promising class of
drugs for the treatment of CD. BACKGROUND: Atopic dermatitis is a chronic inflammatory skin disease
characterized by pruritic skin lesions.
OBJECTIVE: We sought to evaluate the safety and efficacy of multiple doses of
the selective Janus kinase 1 inhibitor upadacitinib in patients with moderate to
severe atopic dermatitis.
METHODS: In the 16-week, double-blind, placebo-controlled, parallel-group,
dose-ranging portion of this 88-week trial in 8 countries (ClinicalTrials.gov,
NCT02925117; ongoing, not recruiting), adults with moderate to severe disease
and inadequate control by topical treatment were randomized 1:1:1:1, using an
interactive response system and stratified geographically, to once-daily
upadacitinib oral monotherapy 7.5, 15, or 30 mg or placebo. The primary end
point was percentage improvement in Eczema Area and Severity Index from baseline
at week 16. Efficacy was analyzed by intention-to-treat in all randomized
patients. Safety was analyzed in all randomized patients who received study
medication, based on actual treatment.
RESULTS: Patients (N = 167) enrolled from November 21, 2016, to April 20, 2017.
All were randomized and analyzed for efficacy (each upadacitinib group, n = 42;
placebo, n = 41); 166 were analyzed for safety (each upadacitinib group, n = 42;
placebo, n = 40). The mean (SE) primary efficacy end point was 39% (6.2%), 62%
(6.1%), and 74% (6.1%) for the upadacitinib 7.5-, 15-, and 30-mg groups,
respectively, versus 23% (6.4%) for placebo (P = .03, <.001, and <.001). Serious
adverse events occurred in 4.8% (2 of 42), 2.4% (1 of 42), and 0% (0 of 42) of
upadacitinib groups (vs 2.5% [1 of 40] for placebo).
CONCLUSIONS: A dose-response relationship was observed for upadacitinib
efficacy; the 30-mg once-daily dose showed the greatest clinical benefit.
Dose-limiting toxicity was not observed. Upadacitinib is a Janus kinase 1 inhibitor developed for treatment of moderate
to severe rheumatoid arthritis (RA) and was recently approved by the US Food and
Drug Administration for this indication in adults who have had an inadequate
response or intolerance to methotrexate. Upadacitinib is currently under
regulatory review by other agencies around the world. Ongoing trials are
investigating the use of upadacitinib in other inflammatory autoimmune diseases.
In this article, we review the clinical pharmacokinetic data available to date
for upadacitinib that supported the clinical development program in RA and
ultimately regulatory applications for upadacitinib in treatment of patients
with moderate to severe RA. BACKGROUND & AIMS: We evaluated the efficacy and safety of upadacitinib, an oral
selective Janus kinase 1 inhibitor, in a randomized trial of patients with
Crohn's disease (CD).
METHODS: We performed a double-blind, phase 2 trial in adults with moderate to
severe CD and inadequate response or intolerance to immunosuppressants or tumor
necrosis factor antagonists. Patients were randomly assigned (1:1:1:1:1:1) to
groups given placebo; or 3 mg, 6 mg, 12 mg, or 24 mg upadacitinib twice daily;
or 24 mg upadacitinib once daily and were evaluated by ileocolonoscopy at weeks
12 or 16 of the induction period. Patients who completed week 16 were
re-randomized to a 36-week period of maintece therapy with upadacitinib. The
primary endpoints were clinical remission at week 16 and endoscopic remission at
week 12 or 16 using the multiple comparison procedure and modeling and the
Cochran-Mantel-Haenszel test, with a 2-sided level of 10%.
RESULTS: Among the 220 patients in the study, clinical remission was achieved by
13% of patients receiving 3 mg upadacitinib, 27% of patients receiving 6 mg
upadacitinib (P < .1 vs placebo), 11% of patients receiving 12 mg upadacitinib,
and 22% of patients receiving 24 mg upadacitinib twice daily, and by 14% of
patients receiving 24 mg upadacitinib once daily, vs 11% of patients receiving
placebo. Endoscopic remission was achieved by 10% (P < .1 vs placebo), 8%, 8%
(P < .1 vs placebo), 22% (P < .01 vs placebo), and 14% (P < .05 vs placebo) of
patients receiving upadacitinib, respectively, vs none of the patients receiving
placebo. Endoscopic but not clinical remission increased with dose during the
induction period. Efficacy was maintained for most endpoints through week 52.
During the induction period, patients in the upadacitinib groups had higher
incidences of infections and serious infections vs placebo. Patients in the
twice-daily 12 mg and 24 mg upadacitinib groups had significant increases in
total, high-density lipoprotein, and low-density lipoprotein cholesterol levels
compared with patients in the placebo group.
CONCLUSIONS: In a phase 2 trial of patients with CD, upadacitinib induced
endoscopic remission in a significant proportion of patients compared with
placebo. Upadacitinib's benefit/risk profile supports further development for
treatment of CD. (Clinicaltrials.gov, Number: NCT02365649). Rheumatoid arthritis (RA) is a chronic inflammatory disease primarily affecting
the joints and is associated with significant levels of disability and reduced
quality of life. Janus kinase (JAK) inhibitors are a relatively new class of
small molecule oral treatments and offer an alternative for patients with RA who
do not respond to conventional or biologic therapy. Upadacitinib is a JAK
inhibitor engineered to be selective for JAK1, and has recently been approved
for use in patients with moderate-to-severe RA. The purpose of this article is
to provide a comprehensive review of upadacitinib, including preclinical
development and characterization, phase I and II studies, and the phase III
SELECT program. Ongoing trials of upadacitinib in additional indications,
including spondyloarthritis, inflammatory bowel disease, and atopic dermatitis,
are also discussed. This phase 1 study characterized the effect of multiple doses of upadacitinib,
an oral Janus kinase 1 selective inhibitor, on the pharmacokinetics of the
cytochrome P450 (CYP) 2B6 substrate bupropion. Healthy subjects (n = 22)
received a single oral dose of bupropion 150 mg alone (study period 1) and on
day 12 of a 16-day regimen of upadacitinib 30 mg once daily (study period 2).
Serial blood samples for measurement of bupropion and hydroxybupropion plasma
concentrations were collected in each study period. The central values (90%
confidence intervals) for the ratios of change were 0.87 (0.79-0.96) for
bupropion maximum plasma concentration (Cmax ), 0.92 (0.87-0.98) for bupropion
area under the plasma-concentration time curve from time 0 to infinity (AUCinf
), 0.78 (0.72-0.85) for hydroxybupropion Cmax , and 0.72 (0.67-0.78) for
hydroxybupropion AUCinf when administered with, relative to when administered
without, upadacitinib. After multiple-dose administration of upadacitinib 30 mg
once daily, upadacitinib mean ± SD AUC0-24 was 641 ± 177 ng·h/mL, and Cmax was
83.3 ± 30.7 ng/mL. These results confirm that upadacitinib has no relevant
effect on pharmacokinetics of substrates metabolized by CYP2B6. Atopic dermatitis is a common, chronic, immune-mediated disease associated with
several comorbidities. Elevated levels of T helper (Th)2, Th22, and also some
Th1 and Th17 cytokines are found in atopic dermatitis skin lesions. Similar to
psoriasis, there is a tendency towards increased use of more targeted therapies.
However, there are still several unmet needs in the treatment of atopic
dermatitis concerning long-term efficacy, tolerability, safety, route of
administration, and cost. The increased knowledge of atopic dermatitis
pathogenesis and the role of Janus kinase/signal transducer and activator of
transcription (JAK/STAT) pathways has allowed the development of new compounds
to inhibit this intracellular signaling pathway implicated in atopic
dermatitis-related immune responses. Currently, JAK inhibitors are an important
focus of therapeutic research for atopic dermatitis. Upadacitinib and
abrocitinib are oral small molecules that inhibit the JAK/STAT pathway by
selectively blocking JAK1. Data from phase II and III trials are encouraging,
revealing that JAK1 inhibitors are effective and well-tolerated agents for
moderate-to-severe atopic dermatitis. Selective JAK1 inhibitors may represent an
important therapeutic option to be included in the treatment algorithm of atopic
dermatitis, owing to oral administration and a favorable safety and tolerability
profile. In this article, we review the current evidence on the efficacy and
safety of oral selective JAK1 inhibitors for the treatment of atopic dermatitis. |
What is known about natriuretic peptide receptor A? | Atrial natriuretic peptide (ANP) and its natriuretic peptide receptors A (NPR-A) and C (NPR-C) are involved in the regulation of physiological and pathophysiological process of blood pressure.
The natriuretic peptide receptor A (NPRA), also known as NPR1 or guanylyl cyclase A, binds ANP and BNP to initiate transmembrane signal transduction by elevating the intracellular levels of cyclic guanosine monophosphate. | BACKGROUND: Atrial natriuretic peptide (ANP) and its natriuretic peptide
receptors A (NPR-A) and C (NPR-C) are involved in the regulation of
physiological and pathophysiological process of blood pressure. The present
study aimed to determine the role of NPR-C in the development of salt-sensitive
hypertension.
METHODS: The Dahl salt-sensitive (DS) and salt-resistant (DR) rats were used in
this study. Animals were matched according to their age and weight, and then
placed on either a high-salt (HS, 8%) or a normal-salt (NS, 0.4%) diet for 6
weeks randomly using random number table. The systolic blood pressure (SBP),
plasmatic sodium concentration (PLNa), urinary sodium excretion (UVNa), and
serum creatinine concentration (Scr) were measured. The concentration of ANP in
blood and tissues (heart and kidney) was detected by enzyme-linked immunosorbent
assay. The expression of ANP, NPR-A, and NPR-C in kidney was evaluated with
western blot analysis. Regarding renal redox state, the concentration changes in
malondialdehyde (MDA), lipofuscin, nicotinamide adenine dinucleotide phosphate
(NADPH) oxidase (Nox), and nitric oxide synthase (NOS) in kidney were detected
by a spectrophotometric method. The kidney damage was evaluated using
pathological techniques and the succinodehydrogenase (SDHase) examination.
Furthermore, after an intra-peritoneal injection of C-atrial natriuretic peptide
(ANP)4-23 (C-ANP4-23), an NPR-C receptor agonist, the SBP, biochemical values in
blood and urine, and renal redox state were evaluated. The paired Student's t
test and analysis of variance followed by the Bonferroni test were performed for
statistical analyses of the comparisons between two groups and multiple groups,
respectively.
RESULTS: The baseline SBP in all groups was within the normal range. At the end
of the 6-week experiment, HS diet significantly increased the SBP in DS rats
from 116.63 ± 2.90 mmHg to 162.25 ± 2.15 mmHg (t = -10.213, P < 0.001). The
changes of SBP were not significant in DS rats on an NS diet and DR rats on an
NS diet or on an HS diet (all P > 0.05). The significant increase of PLNa, UVNa,
and Scr related to an HS diet was found in both DS and DR rats (all P < 0.05).
However, significant changes in the concentration (t = -21.915, P < 0.001) and
expression of renal ANP (t = -3.566, P = 0.016) and the expression of renal
NPR-C (t = 5.864, P = 0.002) were only observed in DS hypertensive rats. The
significantly higher desmin immunochemical staining score (t = -5.715,
P = 0.005) and mitochondrial injury score (t = -6.325, P = 0.003) accompanied by
the lower SDHase concentration (t = 3.972, P = 0.017) revealed mitochondrial
pathologic abnormalities in podocytes in DS rats with an HS diet. The distinct
increases of MDA (t = -4.685, P = 0.009), lipofuscin (t = -8.195, P = 0.001),
and Nox (t = -12.733, P < 0.001) but not NOS (t = -0.328, P = 0.764) in kidneys
were also found in DS hypertensive rats. C-ANP4-23 treatment significantly
decreased the SBP induced by HS in DS rats (P < 0.05), which was still higher
than NS groups with the vehicle or C-ANP4-23 treatment (P < 0.05). Moreover, the
HS-induced increase of MDA, lipofuscin, Nox concentrations, and Nox4 expression
in DS rats was significantly attenuated by C-ANP4-23 treatment as compared with
those with HS diet and vehicle injection (all P < 0.05).
CONCLUSIONS: The results indicated that the renal NPR-C might be involved in the
salt-sensitive hypertension through the damage of mitochondria in podocytes and
the reduction of the anti-oxidative function. Hence, C-ANP4-23 might serve as a
therapeutic agent in treating salt-sensitive hypertension. Atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) are
important biological markers and regulators of cardiac function. The natriuretic
peptide receptor A (NPRA), also known as NPR1 or guanylyl cyclase A, binds ANP
and BNP to initiate transmembrane signal transduction by elevating the
intracellular levels of cyclic guanosine monophosphate. However, the effects and
mechanisms downstream of NPRA are largely unknown. The aim of the present study
was to evaluate the changes in the global pattern of mRNA and circular RNA
(circRNA) expression in NPRA‑/‑ and NPRA+/+ myocardium. Differentially expressed
mRNA molecules were characterised using Gene Ontology and Kyoto Encyclopedia of
Genes and Genomes pathway analysis and were found to be primarily related to
metabolic processes. Moreover, circRNA expression was also examined, and a
possible competing endogenous RNA network consisting of circRNA, microRNA
(miRNA), and mRNA molecules was constructed. The results of this study indicated
that NPRA may play a role in cardiac metabolism, which could be mediated by
circRNA through endogenous competition mechanisms. These findings may provide
insight into future characterisation of various ceRNA network pathways. |
What is the mode of action of dexamethasone? | Glucocorticoids like Dexamethasone have a number of modes of action. While these drugs are used to reduce inflammation, Dexamethasone can also induce apoptosis thru initiation of autophagy, activate glucocorticoid receptors in the treatment of uveitic edema, alter gene expression in allergic asthma prevent tachycardia-induced ionic remodeling by reduction of atrial sodium current I(Na), increase gut permeability and suppress inflammation. in addition, Dexamethasone (Dex) can enhance BMP-2-induced osteoblast differentiation and can differentially modulated dendritic cell maturation and TREM1 signaling pathways in GM-CSF-treated and M-CSF-treated monocytes. Dexamethasone can be used for pain management | Reducing reperfusion injury is effective in reducing flap loss after prolonged
ischemia. Anti-inflammatory therapy reduces reperfusion injury in canine cardiac
muscle and ex vivo rat cremaster muscle; however, to date, there are no studies
involving the use of anti-inflammatory agents in ischemic skin flaps. This study
was designed to assess the effects of dexamethasone and indomethacin on the
viability of rat island groin flaps subjected to 10 hours of ischemia. The
ischemic control and the treatment group flaps were subjected to 10 hours of
ischemia by clamping the inferior epigastric vascular pedicle. The treatment
groups received either intravenous dexamethasone or intravenous indomethacin
after the flap vascular pedicles were clamped. Our results showed significant
improvement (p < 0.05, Fisher's exact test) in ischemic flap survival using
dexamethasone. The specific mode of action of dexamethasone was not
investigated; however, its anti-inflammatory effects were most likely
responsible for the improvement of flap survival by suppressing the circulating
neutrophil and decreasing reperfusion injury. Dexamethasone is easily available
for clinical use, and its use should be considered in cases of prolonged
ischemia in skin flaps. Despite the known effectiveness of anti-inflammatory therapy in reducing
reperfusion injury, no studies to date involve the use of anti-inflammatory
therapy in reducing ischemia-reperfusion injury in fasciocutaneous flaps.
Dexamethasone (a phospholipase A2 inhibitor) and specific cyclooxygenase and
lipoxygenase inhibitors (indomethacin and BW755C) were administered to rats with
ischemic island groin (fasciocutaneous) flaps. Significant improvement in
ischemic flap survival was found with dexamethasone and BW755C. The mode of
action of dexamethasone was not specifically investigated in our study; however,
it may suppress neutrophil function and reduce ischemia-reperfusion injury in
its shared ability with BW755C to reduce the formation of leukotrienes.
Dexamethasone could be applied in the clinical setting to reduce ischemic flap
loss by attenuating the systemic inflammatory response to reperfused
ischemic-damaged tissue. In order to evaluate the mode of action of dexamethasone (DEX) on
macrophage-mediated cytotoxicity and to understand its association with nitric
oxide (NO) production, the effect of DEX on macrophage- and spermine
NONOate-mediated cytotoxicity was studied. DEX caused 100% inhibition of
cytotoxicity by LPS- and IFN gamma-activated macrophages whereas it caused only
partial inhibition of NO production. Inhibition of macrophage-mediated
cytotoxicity by DEX was not reversed by supplementation of rTNF alpha. The
partial inhibition of NO production by DEX was due to partial inhibition of iNOS
mRNA expression. Incubation of macrophages with DEX for up to 24 h prior to
activation did not cause further inhibition of NO production. DEX failed to
inhibit NO production if added 6 h after addition of LPS and IFN gamma. Addition
of P815 cells after the onset of NO production resulted in partial restoration
of cytotoxicity in DEX-treated macrophages. Incubation of P815 cells with
spermine NONOate, a synthetic NO donor, resulted in P815 cell lysis, which was
dose-dependent, had a lag phase of 3 h and was blocked by hemoglobin. DEX also
inhibited spermine NONOate-mediated tumor cell lysis, indicating that DEX may
have a protective effect on tumor targets. These results indicate that DEX
inhibits macrophage-mediated cytotoxicity by decreasing NO production and by
inhibiting the cytotoxic effects of NO on the target cells. Glucocorticoids have been shown repeatedly to inhibit the release of prolactin
(PRL) in the rat but their site and mode of action is unknown. In the present
study, we used an in vitro model to examine the requirement for protein
synthesis for dexamethasone to suppress the release of immunoreactive (ir)-PRL
release from the rat pituitary gland. In addition we have performed a series of
in vitro and in vivo experiments to investigate the potential role in this
regard of lipocortin 1 (LC1), a protein shown previously not only to mediate
aspects of the anti-inflammatory and anti-proliferative actions of the
glucocorticoids but also to contribute to the regulatory actions of the steroids
in the brain-neuroendocrine system. In vitro, the release of ir-PRL from rat
anterior pituitary tissue initiated by submaximal concentrations of VIP (10 nM).
TRH (10 nM) or the adenyl cyclase activator forskolin (100 microM) was reduced
significantly (p < 0.01) by preincubation (2 h) of the tissue with dexamethasone
(0.1 microM). By contrast, ir-PRL release evoked by a submaximal concentration
of the L-Ca2+ channel opener BAY K8644 (10 microM) was unaffected by the steroid
although readily antagonised (p < 0.01) by nifedipine (1-100 microM). Exposure
of the pituitary tissue to dexamethasone (0.1 microM) also caused a pronounced
and highly significant increase in de novo protein synthesis, as assessed by the
incorporation of 14C-lysine into the tissue (p < 0.001). This response was
reduced markedly by the inclusion of the RNA and protein synthesis inhibitors,
actinomycin-D (0.5 micrograms/ml) or cycloheximide (1.0 micrograms/ml), in the
incubation medium (p < 0.001), both of which also effectively abrogated (p <
0.01) the dexamethasone-induced inhibition of the release of ir-PRL evoked by
TRH. VIP and forskolin. Lipocortin I was readily detectable by Western blotting
in protein extracts of freshly excised anterior pituitary tissue: a small
proportion of the protein was found to be attached to the outer surface of the
cells where it was retained by a Ca(2+)-dependent mechanism. Exposure of the
tissue in vitro to dexamethasone (0.1 microM) or corticosterone (0.1 microM) but
not 17 beta-oestradiol (0.1 microM) caused a pronounced increase in the amount
of LC1 attached to the outer surface of the cells and concomitant decrease in
the LC1 content of the intracellular LC1 pool. Addition of an N-terminal LC1
fragment. LC11-188 (10 pg-10 ng/ml), to the incubation medium reduced
significantly (p < 0.01) the increases in ir-PRL release induced in vitro by VIP
(10 nM) and forskolin (100 microM). By contrast, at all concentrations tested.
LC11-188 (10 pg-10 ng/ml) failed to influence (p < 0.05) the highly significant
(p < 0.01) ir-PRL response to TRH (10 nM). Similarly, the inhibitory actions of
dexamethasone (0.1 microM) on the release of ir-PRL induced by VIP (10 nM) or
forskolin (100 microM) but not by TRH (10 nM) were substantially reversed (p <
0.01) by a specific monoclonal anti-LC1 antibody while an isotype-matched
control antibody was without effect. In vivo, rats pretreated with either a
polyclonal anti LC1 antiserum (anti-LC1 pAb, 1 ml/day s.c. for 2 days) or a
corresponding volume of non-immune sheep serum (NSS) responded to stress
(laparotomy under ether anaesthesia) with significant (p < 0.05) increases in
the serum ir-PRL concentration. In the NSS-treated group, the ir-PRL response to
stress was effectively inhibited by dexamethasone (100 micrograms/kg i.p.) which
had no effect on the pre-stress serum ir-PRL concentration. By contrast, in rats
pretreated with anti-LC1 pAb dexamethasone failed to block the stress-induced
release of ir-PRL. The results show clearly that the inhibitory actions of
dexamethasone on PRL release are dependent on de novo protein synthesis and
provide novel evidence for the involvement of both LC1-dependent and
LC1-independent mechanisms. Glucocorticoids (GCs) influence a great variety of cellular functions by at
least three important modes of action: the activation (or repression) of genes
controlled by binding sites for the glucocorticoid receptor (GR), the induction
of apoptosis in lymphocytes and the recently discovered cross-talk to other
transcription factors such as NF-kappaB. In this study we systematically
compared various natural and synthetic steroid hormones frequently used as
therapeutic agents on their ability to mediate these three modes of action.
Betamethasone, triamcinolone, dexamethasone and clobetasol turned out to be the
best inducers of gene expression and apoptosis. All GCs including the
antagonistic compound RU486 efficiently reduced NF-kappaB-mediated
transactivation to comparable extents, suggesting that ligand-induced nuclear
localization of the GR is sufficient for transrepression. Glucocorticoid
treatment of cells did not result in elevated IkappaB-alpha expression, but
impaired the tumor necrosis factor (TNF)-alpha-induced degradation of
IkappaB-alpha without affecting DNA binding of NF-kappaB. The structural
requirements for the various functions of glucocorticoids are discussed. Restenosis after stent implantation is mainly characterized by an inflammatory
response to the procedural injury and an intense fibrocellular response
including smooth muscle cell (SMC) proliferation. After angioplasty alone, the
restenosis process also involves thrombus formation and negative remodeling. Due
to the pleiotropic mode of action exerted by glucocorticoids which include
profound anti-inflammatory and immunosuppressive effects, direct inhibition on
SMC proliferation and apoptosis, their potential in the prevention of restenosis
has gained widespread interest. Over the last decade, preclinical and clinical
data have not been able to conclusively document a robust therapeutic effect on
restenosis after angioplasty or stent implantation. Only recently, preclinical
data and limited observations in humans using drug eluting stents for local drug
delivery have suggested beneficial effects of dexamethasone on neointimal
proliferation. Randomized clinical trials using local drug delivery are expected
to start in the near future. In the light of these ongoing developments, this
review summarizes the pathophysiological basis of glucocorticoid action in the
context of restenosis, provides an overview of the animal data available and
discusses the clinical results that have been gathered over the last decade with
particular emphasis on dexamethasone. PURPOSE: Atrial fibrillation (AF) results in tachycardia-induced ionic
remodeling. Pharmacological prevention of tachycardia-induced ionic remodeling
not only with "classical" antiarrhythmics but also with drugs which provide a
basis for some of the pillars of the so-called "upstream" therapy of AF like
corticosteroids or statins has been proposed as a therapeutic strategy. Amongst
other ion currents, atrial sodium current I(Na) and its tachycardia-induced
alterations play an important role in AF pathophysiology. Thus, effects of a
dexamethasone (DT) and atorvastatin treatment (AT) on atrial sodium current
I(Na) and its tachycardia-induced remodeling were studied in a rabbit model.
METHODS: 9 groups with 4 animals were examined. Atrial pacemaker leads were
implanted in all animals. No rapid atrial pacing (600/min) was performed in the
control group but for 24 or 120 hours in the respective pacing groups.
Instrumentation and pacing did not differ from the respective drug groups but an
additional treatment with dexamethasone or atorvastatin (7 days) was performed.
RESULTS: Rapid atrial pacing (RAP, 600/min) reduced I(Na) after 24 hours (≈
-50%) with no further reduction after 120 hours. DT reduced I(Na) (≈ -20%),
current densities in consecutively tachypaced animals did not differ from those
in untreated animals. AT reduced INa similar as RAP, subsequent RAP did not
further diminish I(Na).
CONCLUSIONS: Impact of corticosteroids and statins on INa and its
tachycardia-induced alterations also contribute to the mode of action of these
substances in upstream treatment of atrial fibrillation. Multimodal pain management, combining analgesics with different mode of action
in order to minimize occurrence of side-effects still providing safe and
efficacious pain management after ambulatory surgery has become standard of
care. The combined use of local anaesthesia in order to reduce noxious influx
during the procedure and reduce postoperative pain is strongly recommended
whenever feasible. Providing oral analgesics paracetamol, and none-steroid
anti-inflammatory drugs or selective Cox-II-inhibitors already prior to
induction in order to provide effective therapeutic concentrations at end of
surgery is a simple and easy way to facilitate the recovery. Single iv.
preoperative dose dexamethasone has been shown not only to be effective in
reducing postoperative nausea and vomiting but also to improve recovery reduce
pain and improve satisfaction. Pregabalin may be used in order to further
enhance the recovery and pain management. Inflammation is a physiological process involved in many diseases. Monitoring
proteins involved in regulatory effects may help to improve our understanding of
inflammation. We have analyzed proteome alterations induced in peripheral blood
mononuclear cells (PBMCs) upon inflammatory activation in great detail using
high-resolution mass spectrometry. Moreover, the activated cells were treated
with dexamethasone to investigate their response to this antiphlogistic drug.
From a total of 6886 identified proteins, 469 proteins were significantly
regulated upon inflammatory activation. Data are available via ProteomeXchange
with identifiers PXD001415-23. Most of these proteins were counter-regulated by
dexamethasone, with some exceptions concerning members of the interferon-induced
protein family. To confirm some of these results, we performed targeted MRM
analyses of selected peptides. The inflammation-induced upregulation of proteins
such as IL-1β, IL-6, CXCL2, and GROα was confirmed, however, with strong
quantitative interindividual differences. Furthermore, the inability of
dexamethasone to downregulate inflammation-induced proteins such as PTX3 and
TSG6 was clearly demonstrated. In conclusion, the relation of cell function as
well as drug-induced modulation thereof was successfully mapped to proteomes,
suggesting targeted analysis as a novel and powerful drug evaluation method.
Although most consequences of dexamethasone were found to be compatible with the
expected mode of action, some unexpected but significant observations may be
related to adverse effects. Classical drug assays are often confined to single molecules and targeting
single pathways. However, it is also desirable to investigate the effects of
complex mixtures on complex systems such as living cells including the natural
multitude of signalling pathways. Evidence based on herbal medicine has
motivated us to investigate potential beneficial health effects of Mucor
racemosus (M rac) extracts. Secondary metabolites of M rac were collected using
a good-manufacturing process (GMP) approved production line and a validated
manufacturing process, in order to obtain a stable product termed SyCircue
(National Drug Code USA: 10424-102). Toxicological studies confirmed that this
product does not contain mycotoxins and is non-genotoxic. Potential effects on
inflammatory processes were investigated by treating stimulated cells with M rac
extracts and the effects were compared to the standard anti-inflammatory drug
dexamethasone on the levels of the proteome and metabolome. Using 2D-PAGE,
slight anti-inflammatory effects were observed in primary white blood
mononuclear cells, which were more pronounced in primary human umbilical vein
endothelial cells (HUVECs). Proteome profiling based on nLC-MS/MS analysis of
tryptic digests revealed inhibitory effects of M rac extracts on
pro-inflammatory cytoplasmic mediators and secreted cytokines and chemokines in
these endothelial cells. This finding was confirmed using targeted proteomics,
here treatment of stimulated cells with M rac extracts down-regulated the
secretion of IL-6, IL-8, CXCL5 and GROA significantly. Finally, the modulating
effects of M rac on HUVECs were also confirmed on the level of the metabolome.
Several metabolites displayed significant concentration changes upon treatment
of inflammatory activated HUVECs with the M rac extract, including spermine and
lysophosphatidylcholine acyl C18:0 and sphingomyelin C26:1, while the bulk of
measured metabolites remained unaffected. Interestingly, the effects of M rac
treatment on lipids were orthogonal to the effect of dexamethasone underlining
differences in the overall mode of action. Heterotopic ossification (HO) consists of ectopic cartilage and bone formation
following severe trauma or invasive surgeries, and a genetic form of it
characterizes patients with Fibrodysplasia Ossificans Progressiva (FOP). Recent
mouse studies showed that HO was significantly inhibited by systemic treatment
with a corticosteroid or the retinoic acid receptor γ agonist Palovarotene.
Because these drugs act differently, the data raised intriguing questions
including whether the drugs affected HO via similar means, whether a combination
therapy would be more effective or whether the drugs may hamper each other's
action. To tackle these questions, we used an effective HO mouse model involving
subcutaneous implantation of Matrigel plus rhBMP2, and compared the
effectiveness of prednisone, dexamathaosone, Palovarotene or combination of.
Each corticosteroid and Palovarotene reduced bone formation at max doses, and a
combination therapy elicited similar outcomes without obvious interference.
While Palovarotene had effectively prevented the initial cartilaginous phase of
HO, the steroids appeared to act more on the bony phase. In reporter assays,
dexamethasone and Palovarotene induced transcriptional activity of their
respective GRE or RARE constructs and did not interfere with each other's
pathway. Interestingly, both drugs inhibited the activity of a reporter
construct for the inflammatory mediator NF-κB, particularly in combination. In
good agreement, immunohistochemical analyses showed that both drugs markedly
reduced the number of mast cells and macrophages near and within the ectopic
Matrigel mass and reduced also the number of progenitor cells. In sum,
corticosteroids and Palovarotene appear to block HO via common and distinct
mechanisms. Most importantly, they directly or indirectly inhibit the
recruitment of immune and inflammatory cells present at the affected site, thus
alleviating the effects of key HO instigators. Although glucocorticoids (GCs) are a mainstay in the clinical management of
asthma, the target cells that mediate their therapeutic effects are unknown.
Contrary to our expectation, we found that GC receptor (GR) expression in immune
cells was dispensable for successful therapy of allergic airway inflammation
(AAI) with dexamethasone. Instead, GC treatment was compromised in mice
expressing a defective GR in the nonhematopoietic compartment or selectively
lacking the GR in airway epithelial cells. Further, we found that an intact GR
dimerization interface was a prerequisite for the suppression of AAI and airway
hyperresponsiveness by GCs. Our observation that the ability of dexamethasone to
modulate gene expression in airway epithelial cells coincided with its potency
to resolve AAI supports a crucial role for transcriptional regulation by the GR
in this cell type. Taken together, we identified an unknown mode of GC action in
the treatment of allergic asthma that might help to develop more specific
therapies in the future. Glucocorticoids (GCs) are potent anti-inflammatory drugs whose mode of action is
complex and still debatable. One likely cellular target of GCs are
monocytes/macrophages. The role of GCs in monocyte survival is also debated.
Although both granulocyte macrophage-colony stimulating factor (GM-CSF) and
macrophage-CSF (M-CSF) are important regulators of macrophage lineage functions
including their survival, the former is often associated with proinflammatory
functions while the latter is important in lineage homeostasis. We report here
that the GC, dexamethasone, induces apoptosis in GM-CSF-treated human monocytes
while having no impact on M-CSF-induced monocyte survival. To understand how
GCs, GM-CSF, and M-CSF are regulating monocyte survival and other functions
during inflammation, we firstly examined the transcriptomic changes elicited by
these three agents in human monocytes, either acting alone or in combination.
Transcriptomic and Ingenuity pathway analyses found that dexamethasone
differentially modulated dendritic cell maturation and TREM1 signaling pathways
in GM-CSF-treated and M-CSF-treated monocytes, two pathways known to be
regulated by ERK1/2 activity. These analyses led us to provide evidence that the
GC inhibits ERK1/2 activity selectively in GM-CSF-treated monocytes to induce
apoptosis. It is proposed that this inhibition of ERK1/2 activity leads to
inactivation of p90 ribosomal-S6 kinase and Bad dephosphorylation leading in
turn to enhanced caspase-3 activity and subsequent apoptosis. Furthermore,
pharmacological inhibition of GC receptor activity restored the ERK1/2 signaling
and prevented the GC-induced apoptosis in GM-CSF-treated monocytes. Increased
tissue macrophage numbers, possibly from enhanced survival due to mediators such
as GM-CSF, can correlate with inflammatory disease severity; also reduction in
these numbers can correlate with the therapeutic benefit of a number of agents,
including GCs. We propose that the ERK1/2 signaling pathway promotes survival of
GM-CSF-treated proinflammatory monocytes, which can be selectively targeted by
GCs as a novel mechanism to reduce local monocyte/macrophage numbers and hence
inflammation. The development of novel bioactive biomaterials is urgently needed to meet the
needs of an aging population. Both sulfated hyaluronic acid and dexamethasone
are candidates for the functionalization of bone grafts, as they have been shown
to enhance the differentiation of osteoblasts from bone marrow stromal cells in
vitro and in vivo. However, the underlying mechanisms are not fully understood.
Furthermore, studies combining different approaches to assess synergistic
potentials are rare. In this study, we aim to gain insights into the mode of
action of both sulfated hyaluronic acid and dexamethasone by a comprehensive
analysis of the cellular fraction, released matrix vesicles, and the
extracellular matrix, combining classical biochemical assays with mass
spectrometry-based proteomics, supported by novel bioinformatical computations.
We found elevated differentiation levels for both treatments, which were further
enhanced by a combination of sulfated hyaluronic acid and dexamethasone. Single
treatments revealed specific effects on osteogenic differentiation.
Dexamethasone activates signalling pathways involved in the differentiation of
osteoblasts, for example, CXC-motif chemokine receptor type 4 and
mitogen-activated protein kinases. The effects of sulfated hyaluronic acid were
predomitly linked to an alteration in the composition of the extracellular
matrix, affecting the synthesis, secretion, and/or activity of fibrillary
(fibronectin and thrombospondin-2) and nonfibrillary (transglutaminase-2,
periostin, and lysyloxidase) extracellular matrix components, including
proteases and their inhibitors (matrix metalloproteinase-2, tissue inhibitor of
metalloproteinase-3). The effects were treatment specific, and less additive or
contrary effects were found. Thus, we anticipate that the synergistic action of
the treatment-specific effects is the key driver in elevated osteogenesis. Dexamethasone (Dex), a synthetic glucocorticoid (GC), in feed has been shown to
increase gut permeability via stress-mediated mechanisms, but the exact mode of
action on gut barrier function is not fully understood. Stress has been reported
to alter the profile and virulence of intestinal flora predisposing for
opportunistic disease. This study aimed to evaluate the relationship between
dietary Dex and recoverable intestinal microbial profile in broilers to better
understand mode of action and refine future uses of the model. Three experiments
were conducted that administered Dex-treated feed for one week in conjunction
with the antibiotics BMD (bacitracin methylene disalicylate) or Baytril®
(enrofloxacin) to evaluate if enteric microbial mechanisms were important in
Dex-induced permeability. Serum fluorescein isothiocyanate-dextran (FITC-d) and
bacterial translocation (BT) have been reported to increase after Dex treatment
and were used to assess gut epithelial leakage. Shifts in bacterial profiles
were also measured on selective agar. Combining Dex with BMD or Baytril resulted
in increased (P < 0.05) serum FITC-d versus Dex-only. Additionally, Baytril did
not reduce aerobic BT and bacterial profiles remained similar after Dex. These
results suggest a minimal role of intestinal microbes in Dex-induced changes to
intestinal barrier function. Glucocorticoids (GCs) are widely used to treat acute graft-versus-host disease
(aGvHD) due to their immunosuppressive activity, but they also reduce the
beneficial graft-versus-leukemia (GvL) effect of the allogeneic T cells
contained in the graft. Here, we tested whether aGvHD therapy could be improved
by delivering GCs with the help of inorganic-organic hybrid oparticles
(IOH-NPs) that preferentially target myeloid cells. IOH-NPs containing the GC
betamethasone (BMP-NPs) efficiently reduced morbidity, mortality, and tissue
damage in a totally MHC mismatched mouse model of aGvHD. Therapeutic activity
was lost in mice lacking the GC receptor (GR) in myeloid cells, confirming the
cell type specificity of our approach. BMP-NPs had no relevant systemic activity
but suppressed cytokine and chemokine gene expression locally in the small
intestine, which presumably explains their mode of action. Most importantly,
BMP-NPs delayed the development of an adoptively transferred B cell lymphoma
better than the free drug, although the overall incidence was unaffected. Our
findings thus suggest that employing IOH-NPs could diminish the risk of relapse
associated with GC therapy of aGvHD patients while still allowing to efficiently
ameliorate the disease. Bone morphogenetic protein-2 (BMP-2) is considered one of the most effective and
extensively used growth factors to induce osteoblast differentiation and
accelerate bone regeneration. Dexamethasone (Dex) with suitable dosage can
enhance BMP-2-induced osteoblast differentiation. To strengthen this synergistic
osteoinductive effect, a pH-responsive chitosan-functionalized mesoporous silica
oparticle (chi-MSN) ensemble was fabricated for dual-delivery of BMP-2 and
Dex. The MSNs are prepared by a CTAB-templated sol-gel method, and further
coated by chitosan via the crosslinking of glycidoxypropyltrimethoxysilane
(GPTMS). The small Dex is encapsulated in the mesopores and the large BMP-2 is
incorporated into the chitosan coating. These chi-MSNs can quickly release BMP-2
in a bioactive form and can then be efficiently endocytosed and further realize
a controlled release of Dex with the decreased pH value into/in cells. With the
synergistic action of BMP-2 and Dex outside and inside the cell, this dual
hybrid delivery system can significantly stimulate osteoblast differentiation
and bone regeneration in vitro and in vivo. Together, this dual-delivery
strategy for osteogenic protein delivery may enhance clinical outcomes by
retaining the bioactivity and optimizing the release mode of the drug/protein. |
What is the phenomenon described as "complex coacervation"? | Here, we demonstrate that charge-mediated phase separation, or complex coacervation, of RNAs with cationic peptides can generate simple model liquid organelles capable of reversibly compartmentalizing biomolecules. Impact of macromolecular crowding on RNA/spermine complex coacervation and oligonucleotide compartmentalization. The addition of PEG decreased both the amount of spermine required for phase separation and the coacervation temperature (TC). | Coacervation was defined as the phenomenon in which a colloidal dispersion
separated into colloid-rich (the coacervate), and colloid-poor phases, both with
the same solvent. Complex coacervation covered the situation in which a mixture
of two polymeric polyions with opposite charge separated into liquid dilute and
concentrated phases, in the same solvent, with both phases, at equilibrium,
containing both polyions. Voorn and Overbeek provided the first theoretical
analysis of complex coacervation by applying Flory-Huggins polymer statistics to
model the random mixing of the polyions and their counter ions in solution,
assuming completely random mixing of the polyions in each phase, with the
electrostatic free energy, ΔG(elect), providing the driving force. However,
experimentally complete randomness does not apply: polyion size, heterogeneity,
chain stiffness and charge density (σ) all affect the equilibrium phase
separation and phase concentrations. Moreover, in pauci-disperse systems
multiple phases are often observed. As an alternative, Veis and Aranyi proposed
the formation of charge paired Symmetrical Aggregates (SA) as an initial step,
followed by phase separation driven by the interaction parameter, χ(23),
combining both entropy and enthalpy factors other than the ΔG(elect)
electrostatic term. This two stage path to equilibrium phase separation allows
for understanding and quantifying and modeling the diverse aggregates produced
by interactions between polyampholyte molecules of different charge density, σ,
and intrinsic polyion structure. Complex coacervation is an associative liquid/liquid phase separation resulting
in the formation of two liquid phases: a polymer-rich coacervate phase and a
dilute continuous solvent phase. In the presence of a third liquid phase in the
form of disperse oil droplets, the coacervate phase tends to wet the oil/water
interface. This affinity has long been known and used for the formation of
core/shell capsules. However, while encapsulation by simple or complex
coacervation has been used empirically for decades, there is a lack of a
thorough understanding of the three-phase wetting phenomena that control the
formation of encapsulated, compound droplets and the role of the viscoelasticity
of the biopolymers involved. In this contribution, we review and discuss the
interplay of wetting phenomena and fluid viscoelasticity in coacervate/oil/water
systems from the perspective of colloid chemistry and fluid dynamics, focusing
on aspects of rheology, interfacial tension measurements at the
coacervate/solvent interface, and on the formation and fragmentation of
three-phase compound drops. There has been a resurgence of interest in complex coacervation, a form of
liquid-liquid phase separation (LLPS) in systems of oppositely charged
macroions, but very few reports describe the somewhat anomalous coacervation
between acidic and basic proteins, which occurs under very narrow ranges of
conditions. We sought to identify the roles of equilibrium interprotein
complexes during the coacervation of β-lactoglobulin dimer (BLG2) with
lactoferrin (LF) and found that this LLPS arises specifically from LF(BLG2)2. We
followed the progress of complexation and coacervation as a function of r, the
LF/BLG molar ratio, using turbidity to monitor the degree of coacervation and
proton release and dynamic light scattering (DLS) to assess the stoichiometry
and abundance of complexes. Isothermal titration calorimetry (ITC) showed that
initial complex formation is endothermic, but a large exotherm related to
coacervate formation obscured other regions. On the basis of turbidimetry,
proton release, and DLS, we propose a speciation diagram that presents the
abundance of various complexes as a function of r. Although multiple species
could be simultaneously present, distinct regions could be identified
corresponding to equilibria among particular protein pairs. Biological cells are highly organized, with numerous subcellular compartments.
Phosphorylation has been hypothesized as a means to control the
assembly/disassembly of liquid-like RNA- and protein-rich intracellular bodies,
or liquid organelles, that lack delimiting membranes. Here, we demonstrate that
charge-mediated phase separation, or complex coacervation, of RNAs with cationic
peptides can generate simple model liquid organelles capable of reversibly
compartmentalizing biomolecules. Formation and dissolution of these liquid
bodies was controlled by changes in peptide phosphorylation state using a
kinase/phosphatase enzyme pair. The droplet-generating phase transition
responded to modification of even a single serine residue. Electrostatic
interactions between the short cationic peptides and the much longer polyanionic
RNAs drove phase separation. Coacervates were also formed on silica beads, a
primitive model for localization at specific intracellular sites. This work
supports phosphoregulation of complex coacervation as a viable mechanism for
dynamic intracellular compartmentalization in membraneless organelles. Oppositely charged polymers can undergo the process of complex coacervation,
which refers to a liquid-liquid phase separation driven by electrostatic
attraction. These materials have demonstrated considerable promise as the basis
for complex, self-assembled materials. In this review, we provide a broad
overview of the theoretical tools used to understand the physical properties of
polymeric coacervates. In particular, we discuss historic theories
(Voorn-Overbeek, Random Phase Approximation), and then describe recent
developments in the field (Field Theoretic, Counterion Release, Molecular
Simulation, and Polymer Reference Interaction Site Model methods). We provide
context for these methods, and map out the patchwork of theoretical models that
are used to describe a diverse array of coacervate systems. We use this review
of the literature to clarify a number of important theoretical challenges
remaining in our physical understanding of complex coacervation. We report the effect of neutral macromolecular crowders poly(ethylene glycol)
(PEG) (8 kDa) and Ficoll (70 kDa) on liquid-liquid phase separation in a
polyuridylic acid (polyU)/spermine complex coacervate system. The addition of
PEG decreased both the amount of spermine required for phase separation and the
coacervation temperature (TC). We interpret these effects on phase behavior as
arising due to excluded volume and preferential interactions on both the
secondary structure/condensation of spermine-associated polyU molecules and on
the association of soluble polyU/spermine polyelectrolyte complexes to form
coacervate droplets. Examination of coacervates formed in the presence of
fluorescently-labeled PEG or Ficoll crowders indicated that Ficoll is
accumulated while PEG is excluded from the coacervate phase, which provides
further insight into the differences in phase behavior. Crowding agents impact
distribution of a biomolecular solute: partitioning of a fluorescently-labeled
U15 RNA oligomer into the polyU/spermine coacervates was increased approximately
two-fold by 20 wt% Ficoll 70 kDa and by more than two orders of magnitude by 20
wt% PEG 8 kDa. The volume of the coacervate phase decreased in the presence of
crowder relative to a dilute buffer solution. These findings indicate that
potential impacts of macromolecular crowding on phase behavior and solute
partitioning should be considered in model systems for intracellular
membraneless organelles. Complex coacervation is an emerging liquid/liquid phase separation (LLPS)
phenomenon that behaves as a membrane-less organelle in living cells. Yet while
one of the critical factors for complex coacervation is temperature, little
analysis and research has been devoted to the temperature effect on complex
coacervation. Here, we performed a complex coacervation of cationic protamine
and multivalent anions (citrate and tripolyphosphate (TPP)). Both mixtures
(i.e., protamine/citrate and protamine/TPP) underwent coacervation in an aqueous
solution, while a mixture of protamine and sodium chloride did not.
Interestingly, the complex coacervation of protamine and multivalent anions
showed upper critical solution temperature (UCST) behavior, and the coacervation
of protamine and multivalent anions was reversible with solution temperature
changes. The large asymmetry in molecular weight between positively charged
protamine (~4 kDa) and the multivalent anions (<0.4 kDa) and strong
electrostatic interactions between positively charged guanidine residues in
protamine and multivalent anions were likely to contribute to UCST behavior in
this coacervation system. Underwater adhesion represents a huge technological challenge as the presence of
water compromises the performance of most commercially available adhesives.
Inspired by natural organisms, we have designed an adhesive based on complex
coacervation, a liquid-liquid phase separation phenomenon. A complex coacervate
adhesive is formed by mixing oppositely charged polyelectrolytes bearing pendant
thermoresponsive poly(N-isopropylacrylamide) (PNIPAM) chains. The material fully
sets underwater due to a change in the environmental conditions, namely
temperature and ionic strength. In this work, we incorporate silica
oparticles forming a hybrid complex coacervate and investigate the resulting
mechanical properties. An enhancement of the mechanical properties is observed
below the PNIPAM lower critical solution temperature (LCST): this is due to the
formation of PNIPAM-silica junctions, which, after setting, contribute to a
moderate increase in the moduli and in the adhesive properties only when
applying an ionic strength gradient. By contrast, when raising the temperature
above the LCST, the mechanical properties are dominated by the association of
PNIPAM chains and the ofiller incorporation leads to an increased
heterogeneity with the formation of fracture planes at the interface between
areas of different concentrations of oparticles, promoting earlier failure of
the network-an unexpected and noteworthy consequence of this hybrid system. Complex coacervation is an associative, liquid-liquid phase separation that can
occur in solutions of oppositely-charged macromolecular species, such as
proteins, polymers, and colloids. This process results in a coacervate phase,
which is a dense mix of the oppositely-charged components, and a supernatant
phase, which is primarily devoid of these same species. First observed almost a
century ago, coacervates have since found relevance in a wide range of
applications; they are used in personal care and food products, cutting edge
biotechnology, and as a motif for materials design and self-assembly. There has
recently been a renaissance in our understanding of this important class of
material phenomena, bringing the science of coacervation to the forefront of
polymer and colloid science, biophysics, and industrial materials design. In
this review, we describe the emergence of a number of these new research
directions, specifically in the context of polymer-polymer complex coacervates,
which are inspired by a number of key physical and chemical insights and driven
by a diverse range of experimental, theoretical, and computational approaches. Multivalent polyions can undergo complex coacervation, producing membraneless
compartments that accumulate ribozymes and enhance catalysis, and offering a
mechanism for functional prebiotic compartmentalization in the origins of life.
Here, we evaluate the impact of lower, more prebiotically-relevant, polyion
multivalency on the functional performance of coacervates as compartments.
Positively and negatively charged homopeptides with 1-100 residues and adenosine
mono-, di-, and triphosphate nucleotides are used as model polyions.
Polycation/polyanion pairs are tested for coacervation, and resulting
membraneless compartments are analyzed for salt resistance, ability to provide a
distinct internal microenvironment (apparent local pH, RNA partitioning), and
effect on RNA structure formation. We find that coacervates formed by phase
separation of the shorter polyions more effectively generated distinct pH
microenvironments, accumulated RNA, and preserved duplexes than those formed by
longer polyions. Hence, coacervates formed by reduced multivalency polyions are
not only viable as functional compartments for prebiotic chemistries, they can
outperform higher molecular weight analogues. |
Which proteins does RG-7992 target? | BFKB8488A is a bispecific antibody against FGFR1 and KLB. | |
Is there a role for TFII-I in megakaryopoiesis? | Yes. TFII-I acts as a repressor of β-globin gene transcription and is implicated in the differentiation of erythrocytes into megakaryopoiesis. Mutations in exon 2 interfere with the synthesis of the full-length isoform of TF II-I and lead to the production of a shortened isoform, TFII, in erythroid cells. TF2-I has a role in embryonic development and differentiation of all eukaryotes but its physiological function is still unclear. | TFII-I is a ubiquitously expressed transcription factor that positively or
negatively regulates gene expression. TFII-I has been implicated in neuronal and
immunologic diseases as well as in thymic epithelial cancer. Williams-Beuren
Syndrome (WBS) is caused by a large hemizygous deletion on chromosome 7q11.23
which encompasses 26-28 genes, including GTF2I, the human gene encoding TFII-I.
A subset of WBS patients has recently been shown to present with macrocytosis, a
mild anemia characterized by enlarged erythrocytes. We conditionally deleted the
TFII-I/Gtf2i gene in adult mice by tamoxifen induced Cre-recombination. Bone
marrow cells revealed defects in erythro-megakaryopoiesis and an increase in
expression of the adult β-globin gene. The data show that TFII-I acts as a
repressor of β-globin gene transcription and that it is implicated in the
differentiation of erythro-megakaryocytic cells. |
Which drugs are included in the VAC regiment for Ewing's sarcoma? | VAC regiment for Ewing's sarcoma includes vincristine, actinomycin, cyclophosphamide. | Vincristine, actinomycin D, and cyclophosphamide (VAC) were administered to 14
patients with Ewing's sarcoma. The primary tumors were treated with radiation
therapy and concurrent chemotherapy. Nine patients had no visible metastases at
diagnosis: two died following the development of pulmonary metastases and the
rest have been free of disease for periods varying from 4 months to 4 1/2 years
following completion of treatment. This contrasts with a 27% survival in
patients previously treated at this center with single agent chemotherapy. Five
other patients had demonstrable metastases at diagnosis: VAC chemotherapy
achieved complete regression of pulmonary metastases in three for 9, 9+ and 24+
months, respectively. Following disappearance of tumor in the latter two,
pulmonary irradiation was administered in an attempt to consolidate the
response, but tumor recurred 6 months later. These patients eventually died of
widespread disease although survival appeared prolonged in comparison to that
seen in past experience. Chemotherapy was well tolerated, although three
patients developed hemorrhagic cystitis, necessitating discontinuation of
cyclophosphamide. The data suggest the potential for prolonged control and an
increase in the cure rate with this therapeutic approach. Soft tissue sarcomas of the paraspinal region comprised 3.3% (56 of 1,688) of
the patients entered and eligible on Intergroup Rhabdomyosarcoma Studies I
(IRS-I) and II (IRS-II) (1972 to 1984). These lesions tended to be greater than
5 cm in diameter at diagnosis, invaded the spinal extradural space, and were of
the extraosseous Ewing's sarcoma or undifferentiated sarcoma subtype in 55% (30
of 56) of the cases. Patients with tumors in clinical groups II, III, and IV
were treated with radiotherapy (XRT) and vincristine-dactinomycin (VA) or VA
plus cyclophosphamide (VAC) +/- doxorubicin. Clinical group I patients treated
on IRS-II did not receive XRT, while those on IRS-I were randomized to receive
VAC +/- XRT. Forty-four of the paraspinal patients (79%) achieved a complete
response (CR) compared with 77% (1,260 of 1,632) for patients with disease in
other sites. Twenty-seven patients (55%) subsequently relapsed (five local,
three regional, four local and distant, and 14 distant). The proportion of
patients surviving 5 years by clinical group (stage) from I to IV were 50%, 50%,
62%, and 27%, respectively. Paraspinal patients had somewhat poorer survival
than patients with disease in other sites, both in IRS-I and IRS-II; the
percentage of paraspinal patients surviving 5 years was 50% and 52% for IRS-I
and IRS-II, respectively, whereas these percentages were 55% and 63% for
patients with disease in other sites. Histology did not influence the CR rate,
but unexpectedly, patients who had embryonal rhabdomyosarcoma (RMS) had the
poorest overall survival rate. We concluded that patients with paraspinal
lesions may require extended-field radiation therapy to reduce the high local
failure rate and more intensive chemotherapy to achieve better local and
systemic tumor control. Phase II studies using ifosfamide both alone and combined with vindesine and
cisplatin have shown the effectiveness of this drug in patients with Ewing's
sarcoma (ES) who had relapsed during VAC (vincristine, actinomycin,
cyclosphosphamide)/VAd (vincristine, Adriamycin) therapy. In November 1984,
these results led the SFOP to adopt a protocol consisting of (1) initial
chemotherapy with three cycles of IVA (ifosfamide, 3 g/m2 on days 1 and 2;
actinomycin D, 750 mg/m2 on days 1-3; vincristine, 1.5 mg/m2 on day 1)
alternating every 3 weeks with IVAd (vincristine on day 22; ifosfamide on days
21-23; Adriamycin, 60 mg/m2 on day 22); (2) radical surgery if possible; (3)
local radiotherapy (RT); and (4) maintece chemotherapy with alternating IVA
and VAd (vincristine, Adriamycin) for up to 9 months. In May 1987, 87 patients
with previously untreated ES entered the study; 61 had localized ES. To date, 54
patients with localized disease and 22 with metastatic disease have finished
initial chemotherapy; 40 patients with localized disease have been evaluated. In
all, 28 patients (70%) were in complete remission (17 patients) or had a tumor
regression of greater than 50% 11 patients) and were considered to be good
responders; 12 patients were considered to be poor responders. After local
radiotherapy in all but 7 patients and surgical resection in 29, 52 of 54 were
considered to be in clinical remission. A total of 13 patients with metastatic
disease were good responders at the completion of the initial chemotherapy.
These results confirm the efficacy of primary chemotherapy using ifosfamide for
the treatment of ES. A randomized study of 264 children and adults with previously untreated
localized Ewing's sarcoma of bone was undertaken between 1973 and 1978 by 83
institutions of three national study groups: Children's Cancer Study Group,
Southwest Oncology Group, and Cancer and Leukemia Group B. The Intergroup Study
was designed to determine if the addition of adriamycin (ADR) or bilateral
pulmonary radiotherapy (RT) to vincristine, dactinomycin, and cyclophosphamide
(VAC therapy) would improve survival and reduce local recurrences and
metastases. All patients received RT to the primary lesion, and the survival
rate after 3 years was 65%. The most effective treatment regimen was VAC plus
ADR; 74% of the patients were free of disease at 2 years. The lengths of
disease-free status and survival of patients treated with VAC plus ADR or VAC
plus RT did not differ. However, both regimens were significantly superior to
treatment with VAC alone. The addition of ADR or bilateral pulmonary RT to VAC
was highly advantageous to patients with nonpelvic primaries. Bone and lung were
the major sites of distant relapse, but the addition of bilateral pulmonary RT
showed no advantage over that of ADR in reducing the occurrence of lung
metastases. These recent results should eliminate some of the pessimism that has
accompanied a diagnosis of Ewing's sarcoma, although distant metastases
continued to be a major reason for failure in the control of this tumor.
Survival of these patients can be improved through well-controlled clinical
trials designed to determine optimal adjuvant chemotherapy and treatment of the
primary lesion. PURPOSE: One hundred thirty of 2,792 patients (5%) registered on three
Intergroup Rhabdomyosarcoma Study clinical trials (IRS-I, -II, and -III) from
1972 to 1991 had an extraosseous Ewing's sarcoma (EOE). We report here the
results of multimodality therapy for this tumor.
PATIENTS AND METHODS: The 130 patients were less than 21 years of age; 70 (54%)
were males. Primary tumor sites were on the trunk in 41 patients, an extremity
in 34, the head/neck in 23, the retroperitoneum/pelvis in 21, and other sites in
11. One hundred fourteen patients had no metastases at diagnosis. In 21
patients, the tumor was completely resected; in 30, the localized or regional
tumor was grossly resected, and in 63 patients, grossly visible sarcoma was left
behind. Sixteen patients (12%) had distant metastases at diagnosis. All patients
were given multiagent chemotherapy and most received irradiation (XRT); none
were treated with bone marrow transplantation.
RESULTS: One hundred seven patients (82%) achieved a complete response. At 10
years, 62%, 61%, and 77% of the patients were alive after treatment on IRS-I,
IRS-II, or IRS-III therapeutic protocols, respectively, similar to figures
obtained in all IRS patients. At last follow-up evaluation, 42 patients had died
of progressive tumor and one of infection. Survival at 10 years was most likely
for patients with tumor that arose in the head and neck, extremities, and trunk,
and for those who underwent grossly complete tumor removal before initiation of
chemotherapy. For patients with localized, gross residual tumor, adding
doxorubicin (DOX) to the combination of vincristine, dactinomycin,
cyclophosphamide (VAC), and XRT did not significantly improve survival in 39
patients (62% alive at 10 years) compared with that of 24 patients treated with
VAC and XRT without DOX (65% alive at 10 years, P = .93).
CONCLUSION: This series indicated that EOE in children is similar to
rhabdomyosarcoma (RMS) in its response to multimodal treatment. No benefit was
apparent from the addition of DOX to VAC chemotherapy in patients with gross
residual EOE. BACKGROUND: Older age and axial location of Ewing's sarcoma have been reported
as unfavorable prognostic factors.
METHODS: The records of patients older than 15 years with the Ewing's family of
tumors were reviewed retrospectively. After the induction chemotherapy
consisting of alternating vincristine, adriablastin, cyclophosphamide (VAC) and
etoposide, ifosfamide with mesna protection (IE), a local treatment modality was
chosen based on tumor and patient characteristics.
RESULTS: Twenty-five patients with a median age of 19 years were evaluated.
Median follow-up was 26 months (range 4-58). Seventeen patients (68%) had died.
In univariate analysis, factors predictive of shorter survival were the patients
presenting with metastatic disease, with the primary tumor located at the
pelvis, those who never achieved complete response to chemotherapy and those who
had chemotherapy for <12 months. Only a negative link with pelvic location was
observed in multivariate analysis [risk ratio 7.5; 95% confidence interval (CI)
1.52-37.06; P = 0.0134]. Median progression-free survival (PFS) and overall
survival (OS) were 10 months (95% CI 6.2-13.8) and 14 months (95% CI 9.3-18.7),
respectively. Cumulative 2-year PFS and OS were 19.0% (95% CI, SD +/-8.4) and
32.7% (95% CI, SD +/-9.8), respectively.
CONCLUSIONS: The prognosis of patients with axial Ewing's sarcoma is dismal
despite an intensive, multimodality approach including multiagent, alternating
chemotherapy, surgery and/or radiotherapy. A more aggressive approach should be
considered for this group of Ewing's sarcoma patients. Files of 133 children with Ewing sarcoma (median age 10 years) were reviewed.
Frequent primary sites were extremities, trunk, pelvis, and cranium. Half of 43
patients with metastases had disease in the lungs. Ten-year overall and
event-free survival rates were 31% and 19%, respectively. Five-year overall
survival rates were 42% in localized and 15% in metastatic disease (p < .0001);
66% in cases with primary tumors < 8 cm and 29% in larger tumors (p = .013). VAC
(vincristine, actinomycin D, and cyclophosphamide) regimens with anthracyclines
resulted in better survival. Presence of distant metastases, large primary
tumors, and pelvic localization were related to poor prognosis. Novel
therapeutic approaches are needed to produce better results, especially in
high-risk patients. BACKGROUND: Filgrastim is an effective granulocyte colony-stimulating factor
(G-CSF) used to reduce periods of neutropenia and the risk of infection after
chemotherapy courses. Pegfilgrastim is a pegylated filgrastim with a longer
plasma half-life that is administered once per cycle.
OBJECTIVE: The aim of this study was to compare the efficacy of pegfilgrastim
and filgrastim administered after chemotherapy in children with Ewing sarcoma.
METHODS: We performed a retrospective chart review of pediatric patients with
Ewing sarcoma. Every patient received both types of G-CSF in different treatment
courses of chemotherapy, which consisted of vincristine, ifosfamide,
doxorubicin, and etoposide (VIDE); vincristine, actinomycin D, and ifosfamide
(VAI); or vincristine, actinomycin D, and cyclophosphamide (VAC). A single
injection of pegfilgrastim 100 microg/kg SC or a daily injection of filgrastim 5
to 10 microg/kg SC was administered 48 to 72 hours after the completion of
chemotherapy. The following data were collected from the medical charts:
proportion of chemotherapy courses with grade 4 neutropenia, duration of grade 4
neutropenia, proportion with severe neutropenia, duration of severe neutropenia,
proportion with febrile neutropenia, duration of antibiotic treatment, duration
of hospitalization, and percentage of patients receiving transfusion. Grade 4
neutropenia was defined as an absolute neutrophil count of <500 x 10(9)/L;
severe neutropenia was defined as a count of <200 x 10(9)/L. Adverse events were
collected from the medical charts.
RESULTS: Twenty children were included (13 girls and 7 boys). The patients'
median age was 12.8 years (range, 9-17 years) and median weight was 45.2 kg
(range, 28-90 kg). A total of 178 chemotherapy courses (108 VIDE; 70 VAI or VAC)
were administered and evaluated, including 134 courses with pegfilgrastim and 44
courses with filgrastim. Considering all types of chemotherapy combined, those
courses in which pegfilgrastim was used were associated with a significantly
lower incidence of severe neutropenia (0.21 vs 0.85; P = 0.03), a shorter
duration of severe neutropenia (0.49 vs 2.36 days; P = 0.01), and a shorter
duration of antibiotic treatment (1.07 vs 4.22 days; P = 0.03) compared with
courses treated with filgrastim. No statistically significant differences were
observed for the proportion and duration of grade 4 neutropenia, proportion of
febrile neutropenia, duration of hospitalization, or red blood cell and platelet
transfusions. Adverse effects were few and comparable between pegfilgrastim and
filgrastim.
CONCLUSIONS: In this retrospective chart review of children with Ewing sarcoma,
using pegfilgrastim after chemotherapy courses was associated with significantly
reduced frequency and shorter duration of severe neutropenia compared with those
courses followed by filgrastim. Randomized controlled trials are needed to
confirm these preliminary observations. Collaborators: Urban Ch, Meister B, Fink FM, Kerbl R, Stollinger O, Schmitt K,
Ebetsberger G, Jones N, Ladenstein R, Gadner H, Maes P, Brichard B, Mazzeo F,
Gil T, Dhooge C, Vermorken JB, Klein A, Kruseova J, Krarup-Hansen A, Nielsen O,
Capra M, Pautard B, Rialland X, Maillart P, Plouvier E, Vérité C, Bui N'guyen B,
Boutard P, Delcambre C, Bay JO, Demeocq F, Couillault G, Isambert N, Plantaz D,
Desfachelles AS, Piguet C, Marec-Bérard P, Blay JY, Gentet JC, Duffaud F,
Bertucci F, Sirvent N, Schmitt C, Rios M, Corradini N, Rolland F, Deville A,
Thyss A, Michon J, Tabone MD, Pierga JY, Millot F, Munzer M, Edan C, Vannier JP,
Guillemet C, Brun E, Berger C, Castex MP, Roche H, Lejars O, Linassier C,
Oberlin O, Le Cesne A, Bauer S, Ebeling P, Flasshove M, Hartmann JT, Reichardt
P, Mertens R, Osieka R, Gnekow A, Schlimok G, Henze G, Thuss-Patience P,
Wickmann L, Reichardt P, Potenberg J, Reichardt P, Bode U, Schmidt-Wolf IG,
Eberl W, Wolff T, Pekrun A, Hofmann A, Andler W, Schneider D, Lauterbach I,
Zickler P, Göbel U, Borkhardt A, Sauerbrey A, Holter W, Bauer S, Ebeling P,
Flasshove M, Eggert A, Klingebiel T, Niemeyer C, Heinz J, Reiter A, Rummel M,
Runde V, Lakomek M, Trümper L, Beck J, Dölken G, Lindemann HW, Körholz D,
Schmoll HJ, Schneppenheim R, Hossfeld DK, Bokemeyer C, Keles H, Welte K, Klein
C, Kulozik A, Egerer G, Cyran J, Strumberg D, Freier W, Graf N, Pfreundschuh M,
Gruhn B, Leipold A, Bentz M, Wehinger H, Schrappe M, Nolte H, Berthold F, Wolf
J, Sternschulte W, Voelpel S, Frieling T, Moessner J, Selle D, Bucsky D, Bartels
H, Mohren M, Fischer T, Kluba U, Gutjahr P, Dittrich M, Reiter S, Dürken M,
Neubauer A, Beyer J, Christiansen H, Erdlenbruch B, Burdach S, Schmid I, Meyer
zum Büschenfelde C, Peschel C, Oduncu F, Jürgens H, Berdel W, Held H,
Hofmann-Wackersreuther G, Müller H, Peters O, Andreesen R, Krause S, Heits F,
Geib-König R, Kasbohm M, Bürger D, Burghard R, Dickerhoff R, Bielack S,
Feddersen I, Clemens MR, Handgretinger R, Hartmann JT, Debatin KM,
Mayer-Steinacker R, Schoengen A, Schlegel P, Lee V, Chik KW, van den Berg H,
Rodenhuis S, Gelderblom AJ, Hoogerbrugge P, Bökkerink JP, Pieters R, Corbett R,
Österlundh G, Behrendtz M, Holmqvist BM, Hjorth L, Petersen C, Jakobson Å,
Hjalmars U, Ljungman G, Angst R, Kühne T, Paulussen M, Leyvraz S, Rischewski J,
Feldges A, Greiner J, Hess U, Exner GU, Niggli F, Knuth A, King D, McCarthy A,
Henry P, Morland B, Rees H, Nicholson J, Traunecker H, Wallace H, Ronghe M,
Simpson E, Cowie F, White J, Picton S, Lewis I, Leahy M, Stark D, Selby PJ,
Heney D, Pizer B, McDowell H, Michalski A, Whelan J, Chisholm J, Pritchard-Jones
K, Judson I, Bren B, Hale J, Verrill M, Walker D, Sokal M, Wheeler K, Lee V,
Gerrard M, Woll P, Lorigan P, Robinson M, Kohler J. Owing to its rarity, rhabdomyosarcoma of the head and neck (HNRMS) has seldom
been discussed in the literature. As most of the data is based only on the
retrospective experiences of tertiary healthcare centers, there are difficulties
in formulating a standard treatment protocol. Moreover, the disease is poorly
understood at its pathological, genetic, and molecular levels. For instance, 20%
of all histological assessment is inaccurate; even an experienced pathologist
can confuse rhabdomyosarcoma (RMS) with neuroblastoma, Ewing's sarcoma, and
lymphoma. RMS can occur sporadically or in association with genetic syndromes
associated with predisposition to other cancers such as Li-Fraumeni syndrome and
neurofibromatosis type 1 (von Recklinghausen disease). Such associations have a
potential role in future gene therapies but are yet to be fully confirmed.
Currently, chemotherapies are ineffective in advanced or metastatic disease and
there is lack of targeted chemotherapy or biological therapy against RMS. Also,
reported uses of chemotherapy for RMS have not produced reasonable responses in
all cases. Despite numerous molecular and biological studies during the past
three decades, the chemotherapeutic regimen remains unchanged. This vincristine,
actinomycin, cyclophosphamide (VAC) regime, described in Kilman, et al. (1973)
and Koop, et al. (1963), has achieved limited success in controlling the
progression of RMS. Thus, the pathogenesis of RMS remains poorly understood
despite extensive modern trials and more than 30 years of studies exploring the
chemotherapeutic options. This suggests a need to explore surgical options for
managing the disease. Surgery is the single most critical therapy for pediatric
HNRMS. However, very few studies have explored the surgical management of
pediatric HNRMS and there is no standard surgical protocol. The aim of this
review is to explore and address such issues in the hope of maximizing the
number of options available for young patients with HNRMS. BACKGROUND Sarcoma botryoides, known as embryonal rhabdomyosarcoma (ERMS), is a
maligt tumor which arises from embryonic muscle cells. The incidence of ERMS
in the uterine cervix rarely occurs at a very young age. With sufficient
resources, management of this disease is not difficult. However, in limited
resources settings, such as in Indonesia, the situation is more challenging.
This case report aims to highlight the difficulties encountered in diagnosing
and treating patients with sarcoma botryoides. CASE REPORT A 3-year-old female
patient came the outpatient clinic of our hospital with a protruding mass from
her vagina resembling a bunch of grapes which easily bled. She underwent surgery
to remove the mass. After the procedure, she did not return to the hospital for
the recommended adjuvant chemotherapy treatment due to limited funds. Three
months later, she came to the outpatient clinic with the same complaint, despite
smaller size. Due to limited resources, we only evaluated the metastasis using
chest x-ray and did not perform intra-operative biopsy. In the second surgery, a
wide excision with 1-2 cm margin was performed, followed by adjuvant
chemotherapy for 6 series. We achieved a satisfactory outcome in this case, and
18 months after the surgery, the patient was still in remission. CONCLUSIONS
Sarcoma botryoides is a rare maligcy. The effective treatment for sarcoma
botryoides is wide excision with safe margin of 1-2 cm, followed by 6-12 cycles
of vincristine, actinomycin D, and cyclophosphamide (VAC) regiment as an
adjuvant chemotherapy. A family's understanding of the treatment plan is
important to achieve desired outcomes. Even with limited resources, this
maligcy can still be properly treated. |
Is YKL-40 used as a biomarker for Alzheimer's disease? | Yes,
cerebrospinal fluid (CSF) YKL-40 levels were reported to be a promising candidate biomarker of glial inflammation in Alzheimer's disease (AD). | INTRODUCTION: Synaptic damage, axonal neurodegeneration, and neuroinflammation
are common features in Alzheimer's disease (AD), frontotemporal dementia (FTD),
and Creutzfeldt-Jakob disease (CJD).
METHODS: Unicentric cohort of 353 participants included healthy control (HC)
subjects, AD continuum stages, genetic AD and FTD, and FTD and CJD. We measured
cerebrospinal fluid neurofilament light (NF-L), neurogranin (Ng), 14-3-3, and
YKL-40 proteins.
RESULTS: Biomarkers showed differences in HC subjects versus AD, FTD, and CJD.
Disease groups differed between them except AD versus FTD for YKL-40. Only NF-L
differed between all stages within the AD continuum. AD and FTD symptomatic
mutation carriers presented differences with respect to HC subjects. Applying
the AT(N) system, 96% subjects were positive for neurodegeneration if 14-3-3 was
used, 94% if NF-L was used, 62% if Ng was used, and 53% if YKL-40 was used.
DISCUSSION: Biomarkers of synapse and neurodegeneration differentiate HC
subjects from neurodegenerative dementias and between AD, FTD, and CJD. NF-L and
14-3-3 performed similar to total tau when AT(N) system was applied. Recently, cerebrospinal fluid (CSF) YKL-40 levels were reported to be a
promising candidate biomarker of glial inflammation in Alzheimer's disease (AD).
To detect how APOE ε4 affects CSF YKL-40 levels in cognitively normal (CN)
states, mild cognitive impairment (MCI) and AD dementia, data from 35 CN
subjects, 63 patients with MCI, and 11 patients with AD from a cross-sectional
study in the Alzheimer's Disease Neuroimaging Initiative (ADNI) database were
investigated. The results showed that CSF YKL-40 concentrations were increased
in the AD dementia group than in the CN group. CSF YKL-40 levels were higher in
APOE ε4 carriers than in noncarriers with MCI. No statistically significant
difference was found in CSF YKL-40 levels between APOE ε4 carrier and
noncarriers in AD and CN subjects. CSF YKL-40 concentrations were tightly
related to CSF tau and p-tau concentrations in the MCI group. Analysis implied
that APOE ε4 might affect CSF YKL-40 levels in MCI subjects, suggesting a
crucial role of APOE ε4 in neuroinflammation in detecting individuals who might
convert to AD from MCI and, thus, as an effective predictive factor. Neurodegenerative diseases comprise a large number of disorders with high impact
on human health. Neurodegenerative processes are caused by various etiological
factors and differ in their clinical presentation. Neuroinflammation is widely
discussed as both a cause and a consequence in the manifestation of these
disorders. The interplay between the two entities is considered as a major
contributor to the ongoing disease progression. An attentive search and
implementation of new and reliable markers specific for the processes of
inflammation and degeneration is still needed. YKL-40 is a secreted glycoprotein
produced by activated glial cells during neuroinflammation. Neuron-specific
enolase (NSE), expressed mainly by neuronal cells, is a long-standing marker for
neuronal damage. The aim of this review is to summarize, clarify, and evaluate
the potential significance and relationship between YKL-40 and NSE as biomarkers
in the monitoring and prognosis of a set of neurological diseases, such as
Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple
sclerosis. YKL-40 appears to be a more reliable biomarker in neurological
diseases than NSE. The more prominent expression pattern of YKL-40 could be
explained with the more obvious involvement of glial cells in pathological
processes accompanying each neurodegenerative disease, whereas reduced NSE
levels are likely related to low metabolic activity and increased death of
neurons. |
On what chromosome is the gene for "SILVER" coat color found for the domestic cat? | Linkage mapping defined a genomic region for SILVER as a 3.3-Mb region, (95.87-99.21 Mb) on chromosome D2 in the domestic cat. | |
Which lncRNAs are induced by heatshock? | Malat1, papas, long noncoding rnas, circrna, neat1, and mirna are induced by heat shock. | Following the initial discovery of the heat shock RNA omega (hsrω) gene of
Drosophila melanogaster to be non-coding (nc) and also inducible by cell stress,
other stress-inducible long non-coding RNAs (lncRNA) have been described in
diverse organisms. In view of the rapid sequence divergence of lncRNAs, present
knowledge of stress trasncriptome is limited and fragmented. Several known
stress-related lncRNAs, associated with specific nuclear speckled domains or
nucleolus, provide structural base for sequestering diverse
RNA-processing/regulatory proteins. Others have roles in transcriptional or
translational inhibition during stress or in signaling pathways; functions of
several other lncRNAs are not yet known. Most stress-related lncRNAs act
primarily by modulating activity of the proteins to which they bind or by
sequestering specific sets of proteins away from the active pool. A common
emerging theme is that a given lncRNA targets one or more protein/s with key
role/s in the cascade of events triggered by the stress and therefore has a
widespread integrative effect. Since proteins associate with RNA through short
sequence motifs, the overall base sequence of functionally similar ncRNAs is
often not conserved except for specific motifs. The rapid evolvability of ncRNA
sequences provides elegant modules for adaptability to changing environment as
binding of one or the other protein to ncRNA can alter its structure and
functions in distinct ways. Thus the stress-related lncRNAs act as hubs in the
cellular networks to coordinate activities of the members within and between
different networks to maintain cellular homeostasis for survival or to trigger
cell death. The field of non-coding RNA (ncRNA) has expanded over the last decade following
the discoveries of several new classes of regulatory ncRNA. A growing amount of
evidence now indicates that ncRNAs are involved even in the most fundamental of
cellular processes. The heat shock response is no exception as ncRNAs are being
identified as integral components of this process. Although this area of
research is only in its infancy, this article focuses on several classes of
regulatory ncRNA (i.e., miRNA, lncRNA, and circRNA), while summarizing their
activities in mammalian heat shock. We also present an updated model integrating
the traditional heat shock response with the activities of regulatory ncRNA. Our
model expands on the mechanisms for efficient execution of the stress response,
while offering a more comprehensive summary of the major regulators and
responders in heat shock signaling. It is our hope that much of what is
discussed herein may help researchers in integrating the fields of heat shock
and ncRNA in mammals. Genomic studies have revealed that humans possess far fewer protein-encoding
genes than originally predicted. These over-estimates were drawn from the
inherent developmental and stimuli-responsive complexity found in humans and
other mammals, when compared to lower eukaryotic organisms. This left a
conceptual void in many cellular networks, as a new class of functional
molecules was necessary for "fine-tuning" the basic proteomic machinery.
Transcriptomics analyses have determined that the vast majority of the genetic
material is transcribed as noncoding RNA, suggesting that these molecules could
provide the functional diversity initially sought from proteins. Indeed, as
discussed in this review, long noncoding RNAs (lncRNAs), the largest family of
noncoding transcripts, have emerged as common regulators of many cellular
stressors; including heat shock, metabolic deprivation and DNA damage. These
stimuli, while divergent in nature, share some common stress-responsive
pathways, notably inhibition of cell proliferation. This role intrinsically
makes stress-responsive lncRNA regulators potential tumor suppressor or
proto-oncogenic genes. As the list of functional RNA molecules continues to
rapidly expand it is becoming increasingly clear that the significance and
functionality of this family may someday rival that of proteins. This article is
part of a Special Issue entitled: Clues to long noncoding RNA taxonomy1, edited
by Dr. Tetsuro Hirose and Dr. Shinichi Nakagawa. Flavivirus infection causes host cell death by initiation of an unfolded protein
response (UPR). UPR is initiated following activation of three ER-membrane
resident sensors, PERK, IRE1α and ATF6, which are otherwise kept inactive
through association with the ER-chaperone GRP78. Activation precedes cellular
and molecular changes that act to restore homeostasis but might eventually
initiate apoptosis. These changes involve influencing function of multiple genes
by either transcriptional or post-transcriptional or post-translational
mechanisms. Transcriptional control includes expression of transcription factor
cascades, which influence cognate gene expression. Malat1 is a long non-coding
RNA which is over-expressed in many human oncogenic tissues and regulates cell
cycle and survival. In this report, for the first time we show activation of
Malat1 following infection by two flaviviruses, both of which activate the UPR
in host cells. The temporal kinetics of expression was restricted to later time
points. Further, Malat1 was also activated by pharmacological inducer of UPR, to
a similar degree. Using drugs that specifically inhibit or activate the PERK or
IRE1α sensors, we demonstrate that signalling through the PERK axis activates
this expression, through a transcriptional mechanism. To our knowledge, this is
the first report of an UPR pathway regulating the expression of an lncRNA. The long noncoding RNA (lncRNA) NEAT1 (nuclear enriched abundant transcript 1)
is the architectural component of nuclear paraspeckles, and it has recently
gained considerable attention as it is abnormally expressed in pathological
conditions such as cancer and neurodegenerative diseases. NEAT1 and paraspeckle
formation are increased in cells upon exposure to a variety of environmental
stressors and believed to play an important role in cell survival. The present
study was undertaken to further investigate the role of NEAT1 in cellular stress
response pathways. We show that NEAT1 is a novel target gene of heat shock
transcription factor 1 (HSF1) and is up-regulated when the heat shock response
pathway is activated by sulforaphane (SFN) or elevated temperature. HSF1 binds
specifically to a newly identified conserved heat shock element in the NEAT1
promoter. In line with this, SFN induced the formation of NEAT1-containing
paraspeckles via an HSF1-dependent mechanism. HSF1 plays a key role in the
cellular response to proteotoxic stress by promoting the expression of a series
of genes, including those encoding molecular chaperones. We have found that the
expression of HSP70, HSP90, and HSP27 is amplified and sustained during heat
shock in NEAT1-depleted cells compared with control cells, indicating that NEAT1
feeds back via an unknown mechanism to regulate HSF1 activity. This
interrelationship is potentially significant in human diseases such as cancer
and neurodegenerative disorders. |
What is the prevalence of poor metabolizers of CYP2C19 among Southern Asians compared to East Asians? | Southeast Asians exhibit a higher prevalence of CYP2C19-poor metabolisers compared with Caucasians and East Asians. | |
Which R/Bioconductor package has been developed for network-based differential expression analysis? | INDEED is an R/Bioconductor package for network based differential expression analysis. INDEED allows users to construct a sparse network based on partial correlation, and to identify biomolecules that have significant changes both at individual expression and pairwise interaction levels. | |
Can propofol cause green urine? | Yes, propofol can cause green discoloration of urine. It is a rare and benign condition, which occurs when clearance of propofol exceeds the hepatic and extrahepatic elimination. | BACKGROUND: The intragastric balloon is filled with saline and methylene blue
dye, to detect balloon deflation early and prevent bowel obstruction, by
monitoring the patient's urine for changes in color.
METHODS: An intragastric balloon filled with 590 ml of saline plus 10 ml of
methylene blue was endoscopically placed under sedation in a 22-year-old man
with morbid obesity (BMI 42 kg/m2). 3 days later, the patient's urine changed to
dark green, and, suspecting a leaking balloon, endoscopy was repeated under
sedation.
RESULTS: No signs of balloon deflation were seen, and the urine returned to
normal color. The next day, the urine turned green again. 7 days later, the
urine discoloration finally disappeared.
CONCLUSION: Propofol, a sedative commonly used by anesthesiologists during
endoscopic procedures, is known to have several side-effects, and urine
discoloration is one of them, albeit rare. This benign side-effect must be known
to obesity surgeons to avoid pointless medical expenditure, unnecessary balloon
removal and distress for patients and clinicians. We describe a 58-year-old man who developed green urine after operation on a
pressure ulcer. The discolouration disappeared gradually after two days. We
think that the use of methylene blue dye during the revision of the wounds and
the use of the sedative propofol could have caused it. INTRODUCTION: Mild therapeutic hypothermia is an increasingly recognised
treatment option to reduce perihemorrhagic edema in severe intracerebral
hemorrhage.
CASE DESCRIPTION: We report the case of a 77-year old woman with atypical
intracerebral hemorrhage that was treated with mild hypothermia in addition to
osmotic therapy. The patient's urine subsequently showed a green discoloration.
Urine discoloration was completely reversible upon discontinuation of propofol.
DISCUSSION AND EVALUATION: Propofol-related urine discoloration may have been
provoked by hypothermia. Due to the benign nature of this side effect, propofol
should be stopped and gastrointestinal function should be supported.
CONCLUSION: More studies are needed to show a causal role of hypothermia and
related decreased enzymatic function. The change in the colour of urine is a known occurrence in an intensive care
setting and is always a cause of concern to the clinicians who have to
differentiate between benign and pathological causes. Herein, we present a case
of 62-year-old postoperative lady, noticed to be passing green coloured urine
believed to be due to intravenous Propofol administration for induction of
general anaesthesia. The green colour of urine due to Propofol occurs when
clearance of Propofol exceeds hepatic elimination, and extrahepatic elimination
of Propofol occurs. This discolouration of urine is a rare (less than 1% cases)
but a benign side effect of Propofol, which is non-nephrotoxic and gets reversed
after discontinuation of the drug. BACKGROUND: Propofol is a short-acting, intravenous sedative-hypnotic agent that
is widely used for the induction and maintece of general anesthesia and
sedation. An uncommon adverse effect of propofol is green discoloration of the
urine, which has been reported not only under general anesthesia but also with
sedation. Although it is assumed that the phenolic derivatives of propofol can
cause green discoloration of the urine, the actual origin remains unknown. The
aim of this report was to identify the origin of the green discoloration of the
urine using liquid chromatography-mass spectrometry (LC-MS).
CLINICAL FEATURES: The patient, a 51-year-old man, was scheduled for his oral
surgery under general anesthesia using propofol. Postoperatively, the color of
his urine was observed to be green. We compared and analyzed both the green
urine and the normal urine using LC-MS.
CONCLUSION: We experienced a case of a patient with green discoloration of the
urine after general anesthesia using propofol. Although LC-MS analysis showed 2
unique peaks in the green urine at 490 and 590 nm, obvious causes were not
revealed. Methylene blue is used to assess the integrity of the bowel and may cause
self-limiting bluish or greenish hue to the urine. Green urine is also caused by
medications such as propofol and infections such as pseudomonas. Knowledge of
the benign nature of this condition prevents unnecessary consultations and
anxiety. BACKGROUND: Analgesia and sedation are key items in intensive care. Recently
published S3 guidelines specifically address treatment of patients with elevated
intracranial pressure.
METHODS: The Austrian Society of Anesthesiology, Resuscitation and Intensive
Care Medicine carried out an online survey of neurointensive care units in
Austria in order to evaluate the current state of practice in the areas of
analgosedation and delirium management in this high-risk patient group.
RESULTS: The response rate was 88%. Induction of anesthesia in patients with
elevated intracranial pressure is carried out with propofol/fentanyl/rocuronium
in >80% of the intensive care units (ICU), 60% use midazolam, 33.3% use
esketamine, 13.3% use barbiturates and 6.7% use etomidate. For maintece of
analgosedation up to 72 h, propofol is used by 80% of the ICUs, followed by
remifentanil (46.7%), sufentanil (40%) and fentanyl (6.7%). For long-term
sedation, 86.7% of ICUs use midazolam, 73.3% sufentanil and 73.3% esketamine.
For sedation periods longer than 7 days, 21.4% of ICUs use propofol. Reasons for
discontinuing propofol are signs of rhabdomyolysis (92.9%), green urine,
elevated liver enzymes (71.4% each) and elevated triglycerides (57.1%). Muscle
relaxants are only used during invasive procedures. Inducing a barbiturate coma
is rated as a last resort by 53.3% of respondents. The monitoring methods used
are bispectral index (BIS™, 61.5% of ICUs), somatosensory-evoked potentials
(SSEP, 53.8%), processed electroencephalography (EEG, 38.5%), intraparenchymal
partial pressure of oxygen (pO2, 38.5%) and microdialysis (23.1%). Sedation and
analgesia are scored using the Richmond agitation and sedation score (RASS,
86.7%), sedation agitation scale (SAS, 6.7%) or numeric rating scale (NRS, 50%)
and behavioral pain scale (BPS, 42.9%), visual analogue scale (VAS), critical
care pain observation tool (CCPOT, each 14.3%) and verbal rating scale (VRS,
7.1%). Delirium monitoring is done using the confusion assessment method for
intensive care units (CAM-ICU, 46.2%) and intensive care delirium screening
checklist (ICDSC, 7.7%). Of the ICUs 46.2% do not carry out delirium monitoring.
CONCLUSION: We found good general compliance with the recommendations of the
current S3 guidelines. Room for improvement exists in monitoring and the use of
scores to detect delirium. The color of urine in patients who receive anesthetic gives much medical
information to a medical team. So, we must check the urine color and know the
cause of discoloration of the urine from anesthetic patients. Green urine is
rare indeed and it is a benign potential side effect of propofol; this
phenomenon is related to the metabolism of propofol. We experienced green urine
from a long-term anesthetized patient who received a continuous infusion of
propofol. We report here on this unusual case and we review the relevant
literature. Propofol is commonly used for induction and maintece of anesthesia, and
sedation in the intensive care unit. In addition, it is also used as an
anesthetic coma treatment for refractory status epilepticus. We present the case
of a 52-year-old man, who developed green urine following propofol coma therapy
for status epilepticus. The urine color recovered following discontinuation of
propofol infusion. The green discoloration of urine is a rare and benign
condition, which occurs when clearance of propofol exceeds the hepatic and
extrahepatic elimination. Green coloured urine is atypical as it usually signifies the presence of an
exogenous substance. Several substances in literature have been associated with
green urine including propofol, biliverdin, metoclopramide, methylene blue,
indigo blue, amitriptyline, methocarbamol, indomethacin, promethazine,
cimetidine and food colourings. We present here a case of middleaged man who
presented to our ER with altered mental status and green coloured urine with
positive urine toxicology reports for benzodiazepine. |
Are Gram positive bacteria able to release extracellular vesicles? | Yes, Gram-negative and Gram-positive bacteria release a variety of membrane vesicles through different formation routes. | Filifactor alocis, a gram-positive, obligate anaerobic rod, is an emerging
periodontal pathogen that is frequently isolated from patients with
periodontitis, peri-implantitis, and apical periodontitis. Recent studies have
shown that extracellular vesicles (EVs) from gram-negative periodontal
pathogens, so-called outer membrane vesicles (OMVs), harbor various effector
molecules responsible for inducing host inflammatory responses. However, there
are no reports of EVs from F. alocis. In this study, we purified and
characterized the protein profiles of EVs from F. alocis and investigated their
immunostimulatory activity on human monocytic THP-1 and human oral keratinocyte
HOK-16B cell lines. Highly pure EVs were obtained from F. alocis using density
gradient ultracentrifugation. Nanoparticle tracking analysis and transmission
electron microscopy showed that F. alocis EVs were between 50 and 270 nm in
diameter. Proteome analysis identified 28 proteins, including lipoproteins,
autolysins, F. alocis complement inhibitor (FACIN), transporter-related
proteins, metabolism-related proteins, and ribosomal proteins. Human cytokine
array analysis showed that F. alocis EVs remarkably induced the expression of
CCL1, CCL2, MIP-1, CCL5, CXCL1, CXCL10, ICAM-1, IL-1β, IL-1ra, IL-6, IL-8, MIF,
SerpinE, and TNF-α in THP-1 cells and CXCL1, G-CSF, GM-CSF, IL-6, and IL-8 in
HOK-16B cells. The immunostimulatory activity of F. alocis EVs was similar to
that of the whole bacterial cells. Our findings provide new insight into the
role of EVs from gram-positive oral bacteria in periodontal diseases. Gram-negative and Gram-positive bacteria release a variety of membrane vesicles
through different formation routes. Knowledge of the structure, molecular cargo
and function of bacterial extracellular vesicles (BEVs) is primarily obtained
from bacteria cultured in laboratory conditions. BEVs in human body fluids have
been less thoroughly investigated most probably due to the methodological
challenges in separating BEVs from their matrix and host-derived eukaryotic
extracellular vesicles (EEVs) such as exosomes and microvesicles. Here, we
present a step-by-step procedure to separate and characterize BEVs from human
body fluids. BEVs are separated through the orthogonal implementation of
ultrafiltration, size-exclusion chromatography (SEC) and density-gradient
centrifugation. Size separates BEVs from bacteria, flagella and cell debris in
stool; and blood cells, high density lipoproteins (HDLs) and soluble proteins in
blood. Density separates BEVs from fibers, protein aggregates and EEVs in stool;
and low-density lipoproteins (LDLs), very-low-density lipoproteins (VLDLs),
chylomicrons, protein aggregates and EEVs in blood. The procedure is label free,
maintains the integrity of BEVs and ensures reproducibility through the use of
automated liquid handlers. Post-separation BEVs are characterized using
orthogonal biochemical endotoxin and Toll-like receptor-based reporter assays in
combination with proteomics, electron microscopy and oparticle tracking
analysis (NTA) to evaluate BEV quality, abundance, structure and molecular
cargo. Separation and characterization of BEVs from body fluids can be done
within 72 h, is compatible with EEV analysis and can be readily adopted by
researchers experienced in basic molecular biology and extracellular vesicle
analysis. We anticipate that this protocol will expand our knowledge on the
biological heterogeneity, molecular cargo and function of BEVs in human body
fluids and steer the development of laboratory research tools and clinical
diagnostic kits. Release of extracellular vesicles (EVs) is a common feature among eukaryotes,
archaea, and bacteria. However, the biogenesis and downstream biological effects
of EVs released from gram-positive bacteria remain poorly characterized. Here,
we report that EVs purified from a community-associated methicillin-resistant
Staphylococcus aureus strain were internalized into human macrophages in vitro
and that this process was blocked by inhibition of the dynamin-dependent
endocytic pathway. Human macrophages responded to S. aureus EVs by TLR2
signaling and activation of NLRP3 inflammasomes through K+ efflux, leading to
the recruitment of ASC and activation of caspase-1. Cleavage of pro-interleukin
(IL)-1β, pro-IL-18, and gasdermin-D by activated caspase-1 resulted in the
cellular release of the mature cytokines IL-1β and IL-18 and induction of
pyroptosis. Consistent with this result, a dose-dependent cytokine response was
detected in the extracellular fluids of mice challenged intraperitoneally with
S. aureus EVs. Pore-forming toxins associated with S. aureus EVs were critical
for NLRP3-dependent caspase-1 activation of human macrophages, but not for TLR2
signaling. In contrast, EV-associated lipoproteins not only mediated TLR2
signaling to initiate the priming step of NLRP3 activation but also modulated EV
biogenesis and the toxin content of EVs, resulting in alterations in IL-1β,
IL-18, and caspase-1 activity. Collectively, our study describes mechanisms by
which S. aureus EVs induce inflammasome activation and reveals an unexpected
role of staphylococcal lipoproteins in EV biogenesis. EVs may serve as a novel
secretory pathway for S. aureus to transport protected cargo in a concentrated
form to host cells during infections to modulate cellular functions. |
Describe a cytokine release syndrome. | The major factor responsible for acute respiratory distress syndrome is the so-called "cytokine storm," which is an aberrant response from the host immune system that induces an exaggerated release of proinflammatory cytokines/chemokines. | Cytokine-release syndrome is a symptom complex associated with the use of many
monoclonal antibodies. Commonly referred to as an infusion reaction, it results
from the release of cytokines from cells targeted by the antibody as well as
immune effector cells recruited to the area. When cytokines are released into
the circulation, systemic symptoms such as fever, nausea, chills, hypotension,
tachycardia, asthenia, headache, rash, scratchy throat, and dyspnea can result.
In most patients, the symptoms are mild to moderate in severity and are managed
easily. However, some patients may experience severe, life-threatening reactions
that result from massive release of cytokines. Severe reactions occur more
commonly during the first infusion in patients with hematologic maligcies who
have not received prior chemotherapy; severe reactions are marked by their rapid
onset and the acuity of associated symptoms. Massive cytokine release is an
oncologic emergency, and special precautions must be taken to prevent
life-threatening complications. This article will present an overview of the
etiology and management of cytokine-release syndrome in patients receiving
monoclonal antibodies to better prepare oncology nurses to safely care for such
patients. Acute cytokine release syndromes are associated with some therapeutic antibodies
in man, leading to a spectrum of clinical signs from nausea, chills and fever to
more serious dose limiting hypotension and tachycardia. When anticipated this
syndrome is typically manageable, however this adverse reaction recently became
headline news when a massive and unexpected cytokine release syndrome occurred
within a few hours of dosing six healthy volunteers with a therapeutic antibody,
putting their lives at risk due to multiple organ failure. Preclinical studies
did not predict this adverse event, emphasising the need to compare the relative
potency of the product in man and the chosen toxicology species, so that
additional margins of safety can be applied when conducting first in man (FIM)
studies if there is uncertainty over the predictability of the toxicology
species. In vitro human PBMC and whole blood cultures may be useful for
predicting cytokine release. However since cytokine release arises through at
least two distinct mechanisms, it should be emphasised that the utility of these
in vitro methods needs to be established for each antibody product. Immune cells secrete small protein molecules that aim for cell-cell
communications. These small molecules are called cytokines. Targeting cancer
cells with administration of bispecific antibodies and natural extracts results
in elevated circulating levels of inflammatory cytokines, including interferon-γ
and interleukin (IL)-6, which lead to cell toxicity. Sustained release of
cytokines due to immunotherapy or hormonal issues causes various diseases. Novel
T cell-engaging therapies and monoclonal antibodies cause cytokine release
syndrome. Efforts are being carried out to maximize the chance for therapeutic
benefit from immunotherapy while minimizing the risk for life-threatening
complications of sustained cytokine release. Neurodegeneration and cardiac
diseases are the prominent diseases caused by inflammatory cytokines. The
phenomenon is called cytokine storm. Cytokines can act antagonistically or
synergistically. Constitutive expression of proinflammatory cytokines such as
IL-3 and IL-6 causes organ damage and unbearable pain. In this review, we will
discuss the regulators of cytokine release, its types, its implications on human
health, and treatment. OBJECTIVES: To describe a pediatric case of cytokine release syndrome secondary
to chimeric antigen receptor-modified T cells associated with acute respiratory
distress syndrome.
DESIGN: Case report.
SETTING: PICU.
PATIENTS: A 14-year-old boy with refractory B cell precursor acute lymphoblastic
leukemia given chimeric antigen receptor cells developed severe cytokine release
syndrome 7 days after the drug product infusion with progressive respiratory
failure. He was admitted to PICU with a clinical picture of acute respiratory
distress syndrome, requiring mechanical ventilation, and secondary
hemophagocytic lymphohistiocytosis.
INTERVENTIONS: Hemoadsorption with cartridge column (Cytosorb) in combination
with continuous renal replacement therapy was associated to the anti-cytokine
therapy (tocilizumab, a monoclonal antibody targeting interleukin-6 receptor).
MEASUREMENTS AND MAIN RESULTS: Decrease of the inflammatory biomarkers
(ferritin, interleukin-6, interleukin-10) in the first 96 hours associated with
a progressive improvement of acute respiratory distress syndrome (Pao2/Fio2
ratio) 7 day after the start of the multimodal treatment.
CONCLUSIONS: This case suggests that hemoadsorption with cartridge column in
combination with continuous renal replacement therapy and tocilizumab is safe
and potentially effective in pediatric patients with severe cytokine release
syndrome. In 2019-2020 a new coronavirus named SARS-CoV-2 was identified as the causative
agent of a several acute respiratory infection named COVID-19, which is causing
a worldwide pandemic. There are still many unresolved questions regarding the
pathogenesis of this disease and especially the reasons underlying the extremely
different clinical course, ranging from asymptomatic forms to severe
manifestations, including the Acute Respiratory Distress Syndrome (ARDS).
SARS-CoV-2 showed phylogenetic similarities to both SARS-CoV and MERS-CoV
viruses, and some of the clinical features are shared between COVID-19 and
previously identified beta-coronavirus infections. Available evidence indicate
that the so called "cytokine storm" an uncontrolled over-production of soluble
markers of inflammation which, in turn, sustain an aberrant systemic
inflammatory response, is a major responsible for the occurrence of ARDS.
Chemokines are low molecular weight proteins with powerful chemoattractant
activity which play a role in the immune cell recruitment during inflammation.
This review will be aimed at providing an overview of the current knowledge on
the involvement of the chemokine/chemokine-receptor system in the cytokine storm
related to SARS-CoV-2 infection. Basic and clinical evidences obtained from
previous SARS and MERS epidemics and available data from COVID-19 will be taken
into account. Cytokine release syndrome is a systemic inflammatory condition that may occur
after treatment with some types of immunotherapy. Adoptive transfer of T cells modified with chimeric antigen receptors (CAR-T
cells) has changed the therapeutic landscape of hematological maligcies,
particularly for acute lymphoblastic leukemia and large B cell lymphoma, where
two different CAR-T products are now considered standard of care. Furthermore,
intense research efforts are under way to expand the clinical application of
CAR-T cell therapy for the benefit of patients suffering from other types of
cancers. Nevertheless, CAR-T cell treatment is associated with toxicities such
as cytokine release syndrome, which can range in severity from mild flu-like
symptoms to life-threatening vasodilatory shock, and a neurological syndrome
termed ICANS (immune effector cell-associated neurotoxicity syndrome), which can
also range in severity from a temporary cognitive deficit lasting only a few
hours to lethal cerebral edema. In this review, we provide an in-depth
discussion of different types of CAR-T cell-associated toxicities, including an
overview of clinical presentation and grading, pathophysiology, and treatment
options. We also address future perspectives and opportunities, with a special
focus on hematological maligcies. In December 2019, a novel coronavirus, COVID-19, was discovered to be the causal
agent of a severe respiratory infection named SARS-CoV-2, and it has since been
recognized worldwide as a pandemic. There are still numerous doubts concerning
its pathogenesis and particularly the underlying causes of the various clinical
courses, ranging from severe manifestations to asymptomatic forms, including
acute respiratory distress syndrome. The major factor responsible for acute
respiratory distress syndrome is the so-called "cytokine storm," which is an
aberrant response from the host immune system that induces an exaggerated
release of proinflammatory cytokines/chemokines. In this review, we will discuss
the role of cytokine storm in COVID-19 and potential treatments with which
counteract this aberrant response, which may be valuable in the clinical
translation. |
Which genes are the main markers of primitive Endoderm (prEN) formation? | The genes involved in primitive endoderm (prEN) formation are fgf4, lrp2, gata4, pdgfra, p dgfrα, gATA6, nanog, pDgfralpha, egam1 and dab2. | The amount of the heterotrimeric G protein subunit G alpha i2 decreases after
the induction of F9 teratocarcinoma cells to become primitive endoderm in the
presence of retinoic acid (RA). The reduction of the G alpha i2 protein in F9
cells by antisense RNA expression was associated with (i) loss of
receptor-mediated inhibition of adenylyl cyclase; (ii) decreased cell doubling
time; (iii) induction of a primitive, endoderm-like phenotype in the absence of
RA; and (iv) production of the differentiation marker tissue-type plasminogen
activator. Expression of a constitutively active, mutant G alpha i2 blocked
RA-induced differentiation. These data suggest the involvement of G alpha i2 in
the control of stem cell differentiation and provide insight into the
involvement of G proteins in growth regulation. F9 embryonal carcinoma (EC) cells were used as a model system to study endoderm
formation during mammalian embryogenesis. F9 cells treated with retinoic acid
(RA) or RA plus dibutyryl cyclic AMP (cAMP) were examined for the expression of
stage-specific embryonic antigen-3 (SSEA-3), a cell surface marker of primitive
and visceral endoderm. SSEA-3 was not detected by indirect immunofluorescence on
the surface of undifferentiated stem cells; however, a subset of SSEA-3-positive
cells appeared with time in culture, amounting to 20% of cells 10 days after
plating. When cultured in the presence of RA, the percentage of SSEA-3-positive
cells increased to 70% of cells 10 days after plating. In contrast, treatment of
cells with RA plus cAMP yielded differentiated cells that were SSEA-3-negative.
These SSEA-3-negative cells exhibited ultrastructural features of parietal yolk
sac endoderm. In contrast, SSEA-3-positive cells appearing in cultures treated
with RA alone exhibited ultrastructural features of primitive endoderm on day 3,
switching to ultrastructural features of parietal endoderm on day 10. Cells with
hybrid features, resembling both visceral and parietal yolk sac, were also seen.
We suggest that differentiation of F9 EC cells into parietal yolk sac-like cells
can occur along two distinct pathways: 1) direct under the combined influence of
RA and cAMP; and 2) indirect, under the influence of RA alone, in which cells
first differentiate into primitive endoderm. Parietal yolk sac-like cells
induced through the latter pathway continue to express SSEA-3, a cell surface
marker of primitive endoderm that is not normally found on parietal endodermal
cells in vivo. Different types of endoderm, including primitive, definitive and mesendoderm,
play a role in the induction and patterning of the vertebrate head. We have
studied the formation of the anterior neural plate in chick embryos using the
homeobox gene GANF as a marker. GANF is first expressed after mesendoderm
ingression from Hensen's node. We found that, after transplantation, neither the
avian hypoblast nor the anterior definitive endoderm is capable of GANF
induction, whereas the mesendoderm (young head process, prechordal plate)
exhibits a strong inductive potential. GANF induction cannot be separated from
the formation of a proper neural plate, which requires an intact lower layer and
the presence of the prechordal mesendoderm. It is inhibited by BMP4 and promoted
by the presence of the BMP antagonist Noggin. In order to investigate the
inductive potential of the mammalian visceral endoderm, we used rabbit embryos
which, in contrast to mouse embryos, allow the morphological recognition of the
prospective anterior pole in the living, pre-primitive-streak embryo. The
anterior visceral endoderm from such rabbit embryos induced neuralization and
independent, ectopic GANF expression domains in the area pellucida or the area
opaca of chick hosts. Thus, the signals for head induction reside in the
anterior visceral endoderm of mammals whereas, in birds and amphibia, they
reside in the prechordal mesendoderm, indicating a heterochronic shift of the
head inductive capacity during the evolution of mammalia. The derivation of the primitive endoderm layer from the pluripotent cells of the
inner cell mass is one of the earliest differentiation and morphogenic events in
embryonic development. GATA4 and GATA6 are the key transcription factors in the
formation of extraembryonic endoderms, but their specific contribution to the
derivation of each endoderm lineage needs clarification. We further analyzed the
dynamic expression and mutant phenotypes of GATA6 in early mouse embryos. GATA6
and GATA4 are both expressed in primitive endoderm cells initially. At embryonic
day (E) 5.0, parietal endoderm cells continue to express both GATA4 and GATA6;
however, visceral endoderm cells express GATA4 but exhibit a reduced expression
of GATA6. By and after E5.5, visceral endoderm cells no longer express GATA6. We
also found that GATA6 null embryos did not form a morphologically recognizable
primitive endoderm layer, and subsequently failed to form visceral and parietal
endoderms. Thus, the current study establishes that GATA6 is essential for the
formation of primitive endoderm, at a much earlier stage then previously
recognized, and expression of GATA6 discriminates parietal endoderm from
visceral endoderm lineages. Gene knockouts in mice have showed that Grb2 and GATA6 are essential for the
formation of primitive endoderm in blastocysts. Here, we confirmed that
implanted Grb2-null blastocysts lack primitive or extraembryonic endoderm cells
either at E4.5 or E5.5 stages. We analyzed the relationship between Grb2 and
GATA6 in the differentiation of embryonic stem (ES) cells to primitive endoderm
in embryoid body models. Upon transfection with GATA6 expression vector,
Grb2-null ES cells underwent endoderm differentiation as indicated by the
expression of the extraembryonic endoderm markers Dab2 and GATA4. Transfection
of GATA4 expression vector also had the same differentiation potency. When
GATA6- or GATA4-transfected Grb2-null ES cells were allowed to aggregate,
fragments of an endoderm layer formed on the surface of the spheroids. The
results suggest that GATA6 is downstream of Grb2 in the inductive signaling
pathway and the expression of GATA6 is sufficient to compensate for the defects
caused by Grb2 deficiency in the development of the primitive and extraembryonic
endoderm. During preimplantation mouse development, the inner cell mass (ICM)
differentiates into two cell lineages--the epiblast and the primitive endoderm
(PrE)--whose precursors are identifiable by reciprocal expression of Nanog and
Gata6, respectively. PrE formation depends on Nanog by a non-cell-autonomous
mechanism. To decipher early cell- and non-cell-autonomous effects, we performed
a mosaic knockdown of Nanog and found that this is sufficient to induce a PrE
fate cell autonomously. Strikingly, in Nanog null embryos, Gata6 expression is
maintained, showing that initiation of the PrE program is Nanog independent.
Treatment of Nanog null embryos with pharmacological inhibitors revealed that
RTK dependency of Gata6 expression is initially direct but later indirect via
Nanog repression. Moreover, we found that subsequent expression of Sox17 and
Gata4--later markers of the PrE--depends on the presence of Fgf4 produced by
Nanog-expressing cells. Thus, our results reveal three distinct phases in the
PrE differentiation program. One of the earliest epithelial-to-mesenchymal transitions in mouse embryogenesis
involves the differentiation of inner cell mass cells into primitive and then
into parietal endoderm. These processes can be recapitulated in vitro using F9
teratocarcinoma cells, which differentiate into primitive endoderm when treated
with retinoic acid (RA) and into parietal endoderm with subsequent treatment
with dibutyryl cyclic adenosine monophosphate (db-cAMP). Our previous work on
how primitive endoderm develops revealed that the Wnt6 gene is upregulated by
RA, leading to the activation of the canonical WNT-β-catenin pathway. The
mechanism by which Wnt6 is regulated was not determined, but in silico analysis
of the human WNT6 promoter region had suggested that the GATA6 and FOXA2
transcription factors might be involved [1]. Subsequent analysis determined that
both Gata6 and Foxa2 mRNA are upregulated in F9 cells treated with RA or RA and
db-cAMP. More specifically, overexpression of Gata6 or Foxa2 alone induced
molecular and morphological markers of primitive endoderm, which occurred
concomitantly with the upregulation of the Wnt6 gene. Gata6- or
Foxa2-overexpressing cells were also found to have increased levels in T-cell
factor (TCF)-dependent transcription, and when these cells were treated with
db-cAMP, they developed into parietal endoderm. Chromatin immunoprecipitation
analysis revealed that GATA6 and FOXA2 were bound to the Wnt6 promoter, and
overexpression studies showed that these transcription factors were sufficient
to switch on the gene expression of a Wnt6 reporter construct. Together, these
results provide evidence for the direct regulation of Wnt6 that leads to the
activation of the canonical WNT-β-catenin pathway and subsequent induction of
primitive extraembryonic endoderm. Embryonic stem (ES) cells have been considered as a valuable renewable source of
materials in regenerative medicine. Recently, we identified the homeoprotein
EGAM1 both in preimplantation mouse embryos and mouse ES cells. Expression of
the Egam1 transcript and its encoded protein was detectable in differentiating
mouse ES cells, while it was almost undetectable in undifferentiated cells. In
the present study, in order to clarify the effect of forced expression of EGAM1
on the differentiation of mouse ES cells in vitro, transfectants expressing
exogenous EGAM1 were generated. Egam1 transfectants promoted differentiation
into cell types expressing Gata6, Gata4, Afp, or Plat, genes associated with
emergence of the extra-embryonic endoderm lineages. On the other hand, Egam1
transfectants inhibited the expression of specific genes for the embryonic
lineages, including Fgf5 (epiblast) and T (mesoderm), in addition to Cdx2, a
specific gene for the extra-embryonic trophectoderm lineages. Changes in the
percentage of cells recognizing by antibodies against specific marker proteins
closely correlated with the expression patterns of their transcripts. Taken
together, the results obtained in this study suggested that mouse ES cells
expressing exogenous EGAM1 preferentially differentiate into extra-embryonic
primitive endoderm lineages, rather than embryonic lineages or extra-embryonic
trophectoderm lineages. Cells of the inner cell mass (ICM) of the mouse blastocyst differentiate into
the pluripotent epiblast or the primitive endoderm (PrE), marked by the
transcription factors NANOG and GATA6, respectively. To investigate the
mechanistic regulation of this process, we applied an unbiased, quantitative,
single-cell-resolution image analysis pipeline to analyze embryos lacking or
exhibiting reduced levels of GATA6. We find that Gata6 mutants exhibit a
complete absence of PrE and demonstrate that GATA6 levels regulate the timing
and speed of lineage commitment within the ICM. Furthermore, we show that GATA6
is necessary for PrE specification by FGF signaling and propose a model where
interactions between NANOG, GATA6, and the FGF/ERK pathway determine ICM cell
fate. This study provides a framework for quantitative analyses of mammalian
embryos and establishes GATA6 as a nodal point in the gene regulatory network
driving ICM lineage specification. A common process during preimplantation mammalian development is blastocyst
formation, which utilizes signaling through fibroblast growth factor receptor 2
(FGFR2), yet the mechanisms through which FGFR2 signaling affect preimplantation
development in bovine embryos remain incompletely understood. Here, we used
RNA-interference to investigate the in vitro development, the frequency of
blastomere apoptosis, and the mRNA expression of developmental marker genes in
FGF receptor 2-knockdown (FGFR2-KD) bovine embryos. A reduction in FGFR2 mRNA
did not affect preimplantation development or the frequency of apoptotic
blastomeres, but did enhanced proliferation of the inner cell mass in
blastocysts (P < 0.05)-which differs from the phenotype reported for bovine
embryos using a pharmacological approach (treatment with the pan-FGFR blocker
PD173074), but agrees with previous results obtained using mouse embryos.
Moreover, the expression of an epiblast marker gene, NANOG, and a primitive
endoderm marker gene, GATA6, remained unchanged, whereas the expression of
another primitive endoderm marker gene, HNF4A, was significantly reduced in
FGFR2-KD embryos. Therefore, FGFR2 signaling appears to be associated with the
regulation of inner cell mass development and proliferation during blastocyst
formation in cattle. Mol. Reprod. Dev. 83: 516-525, 2016. © 2016 Wiley
Periodicals, Inc. |
What are the EMA and FDA recommendations regarding pharmacogenetic testing for abacavir? | Abacavir HSRs are highly associated with the major histocompatibility complex class I. Large studies established the effectiveness of prospective HLA-B*57:01 screening to prevent HSRs to abacavir. Accordingly to these results the abacavir label has been modified: the European Medicines Agency (EMA) and the FDA recommend/suggested that the administration of abacavir must be preceded by a specific genotyping test. The HLA locus is extremely polymorphic, exhibiting many closely related alleles, making it difficult to discriminate HLA-B*57:01 from other related alleles, and a number of different molecular techniques have been developed recently to detect the presence of HLA-B*57:01. | Many pharmacogenomic biomarkers (PGBM) were identified and translated into
clinical practice, affecting the usage of drugs via label updates. In this
context, abacavir is one of the most brilliant examples of pharmacogenetic
studies translated into clinical practice. Pharmacogenetic studies have revealed
that abacavir HSRs are highly associated with the major histocompatibility
complex class I. Large studies established the effectiveness of prospective
HLA-B*57:01 screening to prevent HSRs to abacavir. Accordingly to these results
the abacavir label has been modified: the European Medicines Agency (EMA) and
the FDA recommend/suggested that the administration of abacavir must be preceded
by a specific genotyping test. The HLA locus is extremely polymorphic,
exhibiting many closely related alleles, making it difficult to discriminate
HLA-B*57:01 from other related alleles, and a number of different molecular
techniques have been developed recently to detect the presence of HLA-B*57:01.
In this review, we provide a summary of the available techniques used by
laboratories to genotype HLA-B*57:01, outlining the scientific and
pharmacoeconomics pros and cons. |
Describe PWMScan | PWMScan is used to scan a position weight matrix (PWM) against a genome or, in general, a large set of DNA sequences. The PWM is the most commonly used mathematical model to describe the DNA binding specificity of a transcription factor (TF). | SUMMARY: Transcription factors regulate gene expression by binding to specific
short DNA sequences of 5-20 bp to regulate the rate of transcription of genetic
information from DNA to messenger RNA. We present PWMScan, a fast web-based tool
to scan server-resident genomes for matches to a user-supplied PWM or
transcription factor binding site model from a public database.
AVAILABILITY AND IMPLEMENTATION: The web server and source code are available at
http://ccg.vital-it.ch/pwmscan and https://sourceforge.net/projects/pwmscan,
respectively.
SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics
online. |
Is vocimagene amiretrorepvec effective for glioblastoma? | No. Treatment with vocimagene amiretrorepvec did not improve survival of glioblastoma patients. | PURPOSE: High-grade gliomas (HGGs) are central nervous system tumors with poor
prognoses and limited treatment options. Vocimagene amiretrorepvec (Toca 511) is
a retroviral replicating vector encoding cytosine deaminase, which converts
extended release 5-fluorocytosine (Toca FC) into the anticancer agent,
5-fluorouracil. According to preclinical studies, this therapy kills cancer
cells and immunosuppressive myeloid cells in the tumor microenvironment, leading
to T-cell-mediated antitumor immune activity. Therefore, we sought to elucidate
this immune-related mechanism of action in humans, and to investigate potential
molecular and immunologic indicators of clinical benefit from therapy.
PATIENTS AND METHODS: In a phase I clinical trial (NCT01470794), patients with
recurrent HGG treated with Toca 511 and Toca FC showed improved survival
relative to historical controls, and some had durable complete responses to
therapy. As a part of this trial, we performed whole-exome DNA sequencing,
RNA-sequencing, and multiplex digital ELISA measurements on tumor and blood
samples.
RESULTS: Genetic analyses suggest mutations, copy-number variations, and
neoantigens are linked to survival. Quantities of tumor immune infiltrates
estimated by transcript abundance may potentially predict clinical outcomes.
Peak values of cytokines in peripheral blood samples collected during and after
therapy could indicate response.
CONCLUSIONS: These results support an immune-related mechanism of action for
Toca 511 and Toca FC, and suggest that molecular and immunologic signatures are
related to clinical benefit from treatment. Conflict of interest statement: Conflict of Interest Disclosures: Dr Gruber is a
member of the board for Tocagen Inc. Drs Rao, Hogan, Accomando, Ostertag,
Montellano, Kheoh, and Kabbinavar were Tocagen employees. Dr Cloughesy reported
personal fees from Roche, personal fees from Trizel, personal fees from
Medscape, personal fees from Bayer, personal fees from Amgen, personal fees from
Odonate Therapeutics, personal fees from Pascal Biosciences, personal fees from
Del Mar Pharmaceuticals, personal fees from Tocagen, personal fees from
Kayopharm, personal fees from GW Pharma, personal fees from Kiyatec, personal
fees from AbbVie, personal fees from Boehringer Ingelheim, personal fees from
VBL, personal fees from VBI, personal fees from Deciphera, personal fees from
Agios, personal fees from QED, personal fees from Merck, personal fees from
Genocea, personal fees from Celgene, personal fees from Puma, personal fees from
Lilly, and personal fees from BMS outside the submitted work; in addition, Dr
Cloughesy had a patent to 62/819322 issued and licensed; and Member of the board
for the 501c3 Global Coalition for Adaptive Research and CMO for the entity;
Co-founder and board member of Katmai Pharmaceuticals. Dr Petrecca reported
other from Tocagen during the conduct of the study. Dr Walbert reported personal
fees from Tocagen outside the submitted work. Dr Damek reported grants from
Tocagen during the conduct of the study; grants from NovoCure, grants from Kazia
Therapeutics, grants from Genentech, grants from Orbus, grants from Roche, and
grants from Forma outside the submitted work. Dr Bota reported personal fees
from NovoCure and personal fees from Zai Lab outside the submitted work. Dr
Bettegowda reported he is a consultant for Depuy-Synthes and Bionaut
Pharmaceuticals. The activities associated with those entities are not related
to the work presented in this manuscript. Dr Zhu reported grants from Tocagen,
Inc and personal fees from Tocagen, Inc during the conduct of the study. Dr
Iwamoto reported personal fees from Tocagen during the conduct of the study;
personal fees from Merck, personal fees from Guidepoint, grants from BMS,
personal fees from NovoCure, personal fees from Alexion, personal fees from
AbbVie, and personal fees from Regeneron outside the submitted work. Dr
Placantonakis reported personal fees from Tocagen during the conduct of the
study; personal fees from Monteris, personal fees from Synaptive, and personal
fees from Robeaute outside the submitted work; in addition, Dr Placantonakis had
a patent to “Method to treat high grade glioma” pending. Dr Brem reported
personal fees from Tocagen during the conduct of the study. Dr Piccioni reported
personal fees from Tocagen during the conduct of the study. Dr Chen reported
other from Tocagen during the conduct of the study; personal fees from Tocagen
outside the submitted work. Dr Gruber reported grants from the US Food and Drug
Administration orphan drug grant, other from Apollo Bio, other from Abentis, and
other from Denovo Pharma during the conduct of the study; other from Apollo Bio,
other from Abentis, and other from Denovo pharma outside the submitted work; in
addition, Dr Gruber had a patent to many pending, issued, licensed, and with
royalties paid, a patent to many pending, issued, and licensed, and a patent to
many pending, issued, and licensed; and Stock and option ownership in Tocagen.
Dr Hogan reported other from Tocagen Inc during the conduct of the study. Dr
Accomando reported personal fees from Tocagen Inc. during the conduct of the
study; personal fees from Tocagen Inc. outside the submitted work. Dr Ostertag
reported a patent to US20130130986A1 issued, a patent to US20130323301A1, a
patent to US20180021365A1, and a patent to US20140178340A1 . Dr Montellano
reported grants from the US Food and Drug Administration Office of Orphan
Products Development during the conduct of the study. Dr Kheoh reported other
from Tocagen Inc during the conduct of the study. Dr Kabbinavar reported other
from Tocagen Inc during the conduct of the study; other from Tocagen outside the
submitted work; and Employee of Tocagen Inc. Dr Vogelbaum reported personal fees
and other from Tocagen during the conduct of the study; other from Infuseon
Theraepeutics and personal fees from Celgene outside the submitted work. No
other disclosures were reported. |
List the major royal jelly proteins in Apis mellifera. | The genome of the western honeybee (Apis mellifera) harbors nine transcribed major royal jelly protein genes (mrjp1-9) which originate from a single-copy precursor via gene duplication. | The honey from chestnut, acacia, sunflower, eucalyptus and orange was analysed
for its proteome content, in order to see if any plant proteins present would
allow the proteo-typing of these different varieties. Since the total protein
content turned out to be minute, 200g of each honey type were diluted to 1L and
then added with ProteoMiner to enhance the visibility of the proteinaceous
material. All bands visible in the SDS-PAGE profile of each type of honey were
eluted, digested and identified by mass spectrometry in a LTQ-XL instrument. It
turned out that all proteins identified (except one, the enzyme
glyceraldehyde-3-phosphate dehydrogenase from Mesembryanthemum crystallinum)
were not of plant origin but belonged to the Apis mellifera proteome. Among the
total proteins identified (eight, but only seven as basic constituents of all
types of honey) five belonged to the family of major royal jelly proteins 1-5,
and were also the most abundant ones in any type of honey, together with
α-glucosidase and defensin-1. It thus appears that honey has a proteome
resembling the royal jelly proteome (but with considerably fewer species),
except that its protein concentration is lower by three to four orders of
magnitude as compared to royal jelly. Attempts at identifying additional plant
(pollen, nectar) proteins via peptidome analysis were unsuccessful. BACKGROUND: In the honeybee Apis mellifera, female larvae destined to become a
queen are fed with royal jelly, a secretion of the hypopharyngeal glands of
young nurse bees that rear the brood. The protein moiety of royal jelly
comprises mostly major royal jelly proteins (MRJPs) of which the coding genes
(mrjp1-9) have been identified on chromosome 11 in the honeybee's genome.
RESULTS: We determined the expression of mrjp1-9 among the honeybee worker caste
(nurses, foragers) and the sexuals (queens (unmated, mated) and drones) in
various body parts (head, thorax, abdomen). Specific mrjp expression was not
only found in brood rearing nurse bees, but also in foragers and the sexuals.
CONCLUSIONS: The expression of mrjp1 to 7 is characteristic for the heads of
worker bees, with an elevated expression of mrjp1-4 and 7 in nurse bees compared
to foragers. Mrjp5 and 6 were higher in foragers compared to nurses suggesting
functions in addition to those of brood food proteins. Furthermore, the
expression of mrjp9 was high in the heads, thoraces and abdomen of almost all
female bees, suggesting a function irrespective of body section. This completely
different expression profile suggests mrjp9 to code for the most ancestral major
royal jelly protein of the honeybee. The genome of the western honeybee (Apis mellifera) harbors nine transcribed
major royal jelly protein genes (mrjp1-9) which originate from a single-copy
precursor via gene duplication. The first MRJP was identified in royal jelly, a
secretion of the bees' hypopharyngeal glands that is used by young worker bees,
called nurses, to feed developing larvae. Thus, MRJPs are frequently assumed to
mainly have functions for developing bee larvae and to be expressed in the food
glands of nurse bees. In-depth knowledge on caste- and age-specific role and
abundance of MRJPs is missing. We here show, using combined quantitative
real-time PCR with quantitative mass spectrometry, that expression and protein
amount of mrjp1-5 and mrjp7 show an age-dependent pattern in worker's
hypopharyngeal glands as well as in brains, albeit lower relative abundance in
brains than in glands. Expression increases after hatching until the nurse bee
period and is followed by a decrease in older workers that forage for plant
products. Mrjp6 expression deviates considerably from the expression profiles of
the other mrjps, does not significantly vary in the brain, and shows its highest
expression in the hypopharyngeal glands during the forager period. Furthermore,
it is the only mrjp of which transcript abundance does not correlate with
protein amount. Mrjp8 and mrjp9 show, compared to the other mrjps, a very low
expression in both tissues. Albeit mrjp8 mRNA was detected via qPCR, the protein
was not quantified in any of the tissues. Due to the occurrence of MRJP8 and
MRJP9 in other body parts of the bees, for example, the venom gland, they might
not have a hypopharyngeal gland- or brain-specific function but rather functions
in other tissues. Thus, mrjp1-7 but not mrjp8 and mrjp9 might be involved in the
regulation of phenotypic plasticity and age polyethism in worker honeybees. Understanding the effect of pesticides on the survival of honeybee colonies is
important because these pollinators are reportedly declining globally. In the
present study, we examined the changes in the head proteome of nurse honeybees
exposed to individual and combined pesticides (the fungicide pyraclostrobin and
the insecticide fipronil) at field-relevant doses (850 and 2.5 ppb,
respectively). The head proteomes of bees exposed to pesticides were compared
with those of bees that were not exposed, and proteins with differences in
expression were identified by mass spectrometry. The exposure of nurse bees to
pesticides reduced the expression of four of the major royal jelly proteins
(MRJP1, MRJP2, MRJP4, and MRJP5) and also several proteins associated with
carbohydrate metabolism and energy synthesis, the antioxidant system,
detoxification, biosynthesis, amino acid metabolism, transcription and
translation, protein folding and binding, olfaction, and learning and memory.
Overall, when pyraclostrobin and fipronil were combined, the changes in protein
expression were exacerbated. Our results demonstrate that vital proteins and
metabolic processes are impaired in nurse honeybees exposed to pesticides in
doses close to those experienced by these insects in the field, increasing their
susceptibility to stressors and affecting the nutrition and maintece of both
managed and natural colonies. |
What is BEL(Biological Expression Language) used for? | Biological Expression Language (BEL) is a novel method for the statistical extraction of causal relation relationships from large biomedical literature datasets. | Towards the development of a systems biology-based risk assessment approach for
environmental toxicants, including tobacco products in a systems toxicology
setting such as the "21st Century Toxicology", we are building a series of
computable biological network models specific to non-diseased pulmonary and
cardiovascular cells/tissues which capture the molecular events that can be
activated following exposure to environmental toxicants. Here we extend on
previous work and report on the construction and evaluation of a mechanistic
network model focused on DNA damage response and the four main cellular fates
induced by stress: autophagy, apoptosis, necroptosis, and senescence. In total,
the network consists of 34 sub-models containing 1052 unique nodes and 1538
unique edges which are supported by 1231 PubMed-referenced literature citations.
Causal node-edge relationships are described using the Biological Expression
Language (BEL), which allows for the semantic representation of life science
relationships in a computable format. The Network is provided in .XGMML format
and can be viewed using freely available network visualization software, such as
Cytoscape. Neurodegenerative as well as autoimmune diseases have unclear aetiologies, but
an increasing number of evidences report for a combination of genetic and
epigenetic alterations that predispose for the development of disease. This
review examines the major milestones in epigenetics research in the context of
diseases and various computational approaches developed in the last decades to
unravel new epigenetic modifications. However, there are limited studies that
systematically link genetic and epigenetic alterations of DNA to the aetiology
of diseases. In this work, we demonstrate how disease-related epigenetic
knowledge can be systematically captured and integrated with heterogeneous
information into a functional context using Biological Expression Language
(BEL). This novel methodology, based on BEL, enables us to integrate epigenetic
modifications such as DNA methylation or acetylation of histones into a specific
disease network. As an example, we depict the integration of epigenetic and
genetic factors in a functional context specific to Parkinson's disease (PD) and
Multiple Sclerosis (MS). Biological expression language (BEL) is one of the most popular languages to
represent the causal and correlative relationships among biological events.
Automatically extracting and representing biomedical events using BEL can help
biologists quickly survey and understand relevant literature. Recently, many
researchers have shown interest in biomedical event extraction. However, the
task is still a challenge for current systems because of the complexity of
integrating different information extraction tasks such as named entity
recognition (NER), named entity normalization (NEN) and relation extraction into
a single system. In this study, we introduce our BelSmile system, which uses a
semantic-role-labeling (SRL)-based approach to extract the NEs and events for
BEL statements. BelSmile combines our previous NER, NEN and SRL systems. We
evaluate BelSmile using the BioCreative V BEL task dataset. Our system achieved
an F-score of 27.8%, ∼7% higher than the top BioCreative V system. The three
main contributions of this study are (i) an effective pipeline approach to
extract BEL statements, and (ii) a syntactic-based labeler to extract
subject-verb-object tuples. We also implement a web-based version of BelSmile
(iii) that is publicly available at iisrserv.csie.ncu.edu.tw/belsmile. Biological expression language (BEL) is one of the main formal representation
models of biological networks. The primary source of information for curating
biological networks in BEL representation has been literature. It remains a
challenge to identify relevant articles and the corresponding evidence
statements for curating and validating BEL statements. In this paper, we
describe BELTracker, a tool used to retrieve and rank evidence sentences from
PubMed abstracts and full-text articles for a given BEL statement (per the 2015
task requirements of BioCreative V BEL Task). The system is comprised of three
main components, (i) translation of a given BEL statement to an information
retrieval (IR) query, (ii) retrieval of relevant PubMed citations and (iii)
finding and ranking the evidence sentences in those citations. BELTracker uses a
combination of multiple approaches based on traditional IR, machine learning,
and heuristics to accomplish the task. The system identified and ranked at least
one fully relevant evidence sentence in the top 10 retrieved sentences for 72
out of 97 BEL statements in the test set. BELTracker achieved a precision of
0.392, 0.532 and 0.615 when evaluated with three criteria, namely full, relaxed
and context criteria, respectively, by the task organizers. Our team at Mayo
Clinic was the only participant in this task. BELTracker is available as a
RESTful API and is available for public use.Database URL:
http://www.openbionlp.org:8080/BelTracker/finder/Given_BEL_Statement. Automatic extraction of biological network information is one of the most
desired and most complex tasks in biological and medical text mining. Track 4 at
BioCreative V attempts to approach this complexity using fragments of
large-scale manually curated biological networks, represented in Biological
Expression Language (BEL), as training and test data. BEL is an advanced
knowledge representation format which has been designed to be both human
readable and machine processable. The specific goal of track 4 was to evaluate
text mining systems capable of automatically constructing BEL statements from
given evidence text, and of retrieving evidence text for given BEL statements.
Given the complexity of the task, we designed an evaluation methodology which
gives credit to partially correct statements. We identified various levels of
information expressed by BEL statements, such as entities, functions, relations,
and introduced an evaluation framework which rewards systems capable of
delivering useful BEL fragments at each of these levels. The aim of this
evaluation method is to help identify the characteristics of the systems which,
if combined, would be most useful for achieving the overall goal of
automatically constructing causal biological networks from text. Success in extracting biological relationships is mainly dependent on the
complexity of the task as well as the availability of high-quality training
data. Here, we describe the new corpora in the systems biology modeling language
BEL for training and testing biological relationship extraction systems that we
prepared for the BioCreative V BEL track. BEL was designed to capture
relationships not only between proteins or chemicals, but also complex events
such as biological processes or disease states. A BEL opub is the smallest
unit of information and represents a biological relationship with its
provece. In BEL relationships (called BEL statements), the entities are
normalized to defined namespaces mainly derived from public repositories, such
as sequence databases, MeSH or publicly available ontologies. In the BEL
opubs, the BEL statements are associated with citation information and
supportive evidence such as a text excerpt. To enable the training of extraction
tools, we prepared BEL resources and made them available to the community. We
selected a subset of these resources focusing on a reduced set of namespaces,
namely, human and mouse genes, ChEBI chemicals, MeSH diseases and GO biological
processes, as well as relationship types 'increases' and 'decreases'. The
published training corpus contains 11 000 BEL statements from over 6000
supportive text excerpts. For method evaluation, we selected and re-annotated
two smaller subcorpora containing 100 text excerpts. For this re-annotation, the
inter-annotator agreement was measured by the BEL track evaluation environment
and resulted in a maximal F-score of 91.18% for full statement agreement. In
addition, for a set of 100 BEL statements, we do not only provide the gold
standard expert annotations, but also text excerpts pre-selected by two
automated systems. Those text excerpts were evaluated and manually annotated as
true or false supportive in the course of the BioCreative V BEL track
task.Database URL: http://wiki.openbel.org/display/BIOC/Datasets. Network-based approaches have become extremely important in systems biology to
achieve a better understanding of biological mechanisms. For network
representation, the Biological Expression Language (BEL) is well designed to
collate findings from the scientific literature into biological network models.
To facilitate encoding and biocuration of such findings in BEL, a BEL
Information Extraction Workflow (BELIEF) was developed. BELIEF provides a
web-based curation interface, the BELIEF Dashboard, that incorporates text
mining techniques to support the biocurator in the generation of BEL networks.
The underlying UIMA-based text mining pipeline (BELIEF Pipeline) uses several
named entity recognition processes and relationship extraction methods to detect
concepts and BEL relationships in literature. The BELIEF Dashboard allows easy
curation of the automatically generated BEL statements and their context
annotations. Resulting BEL statements and their context annotations can be
syntactically and semantically verified to ensure consistency in the BEL
network. In summary, the workflow supports experts in different stages of
systems biology network building. Based on the BioCreative V BEL track
evaluation, we show that the BELIEF Pipeline automatically extracts
relationships with an F-score of 36.4% and fully correct statements can be
obtained with an F-score of 30.8%. Participation in the BioCreative V
Interactive task (IAT) track with BELIEF revealed a systems usability scale
(SUS) of 67. Considering the complexity of the task for new users-learning BEL,
working with a completely new interface, and performing complex curation-a score
so close to the overall SUS average highlights the usability of BELIEF.Database
URL: BELIEF is available at http://www.scaiview.com/belief/. SUMMARY: Biological Expression Language (BEL) assembles knowledge networks from
biological relations across multiple modes and scales. Here, we present PyBEL; a
software package for parsing, validating, converting, storing, querying, and
visualizing networks encoded in BEL.
AVAILABILITY AND IMPLEMENTATION: PyBEL is implemented in platform-independent,
universal Python code. Its source is distributed under the Apache 2.0 License at
https://github.com/pybel.
CONTACT: [email protected].
SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics
online. The BioCreative-V community proposed a challenging task of automatic extraction
of causal relation network in Biological Expression Language (BEL) from the
biomedical literature. Previous studies on this task largely used models induced
from other related tasks and then transformed intermediate structures to BEL
statements, which left the given training corpus unexplored. To make full use of
the BEL training corpus, in this work, we propose a deep learning-based approach
to extract BEL statements. Specifically, we decompose the problem into two
subtasks: entity relation extraction and entity function detection. First, two
attention-based bidirectional long short-term memory networks models are used to
extract entity relation and entity function, respectively. Then entity relation
and their functions are combined into a BEL statement. In order to boost the
overall performance, a strategy of threshold filtering is applied to improve the
precision of identified entity functions. We evaluate our approach on the
BioCreative-V Track 4 corpus with or without gold entities. The experimental
results show that our method achieves the state-of-the-art performance with an
overall F1-measure of 46.9% in stage 2 and 21.3% in stage 1, respectively. BACKGROUND: Extracting relations between bio-entities from biomedical literature
is often a challenging task and also an essential step towards biomedical
knowledge expansion. The BioCreative community has organized a shared task to
evaluate the robustness of the causal relationship extraction algorithms in
Biological Expression Language (BEL) from biomedical literature.
METHOD: We first map the sentence-level BEL statements in the BC-V training
corpus to the corresponding text segments, thus generating hierarchically tagged
training instances. A hierarchical sequence labeling model was afterwards
induced from these training instances and applied to the test sentences in order
to construct the BEL statements.
RESULTS: The experimental results on extracting BEL statements from BioCreative
V Track 4 test corpus show that our method achieves promising performance with
an overall F-measure of 31.6%. Furthermore, it has the potential to be enhanced
by adopting more advanced machine learning approaches.
CONCLUSION: We propose a framework for hierarchical relation extraction using
hierarchical sequence labeling on the instance-level training corpus derived
from the original sentence-level corpus via word alignment. Its main advantage
is that we can make full use of the original training corpus to induce the
sequence labelers and then apply them to the test corpus. Knowledge of the molecular interactions of biological and chemical entities and
their involvement in biological processes or clinical phenotypes is important
for data interpretation. Unfortunately, this knowledge is mostly embedded in the
literature in such a way that it is unavailable for automated data analysis
procedures. Biological expression language (BEL) is a syntax representation
allowing for the structured representation of a broad range of biological
relationships. It is used in various situations to extract such knowledge and
transform it into BEL networks. To support the tedious and time-intensive
extraction work of curators with automated methods, we developed the BEL track
within the framework of BioCreative Challenges. Within the BEL track, we provide
training data and an evaluation environment to encourage the text mining
community to tackle the automatic extraction of complex BEL relationships. In
2017 BioCreative VI, the 2015 BEL track was repeated with new test data.
Although only minor improvements in text snippet retrieval for given statements
were achieved during this second BEL task iteration, a significant increase of
BEL statement extraction performance from provided sentences could be seen. The
best performing system reached a 32% F-score for the extraction of complete BEL
statements and with the given named entities this increased to 49%. This time,
besides rule-based systems, new methods involving hierarchical sequence labeling
and neural networks were applied for BEL statement extraction. |
Which database contains gene expression data for yeast? | We developed the ExpressDB database for yeast RNA expression data and loaded it with approximately 17.5 million pieces of data reported by 11 studies with three different kinds of high-throughput RNA assays. | We report steps toward the systematic management, standardization, and analysis
of functional genomics data. We developed the ExpressDB database for yeast RNA
expression data and loaded it with approximately 17.5 million pieces of data
reported by 11 studies with three different kinds of high-throughput RNA assays.
A web-based tool supports queries across the data from these studies. We
examined comparability of data by converting data from 9 studies (217
conditions) into mRNA relative abundance estimates (ERAs) and by clustering of
conditions by ERAs. We report on generation of ERAs and condition clustering for
non-microarray data (5 studies, 63 conditions) and describe initial attempts to
generate microarray-based ERAs (4 studies, 154 conditions), which exhibit
increased error, on our web site http://arep.med.harvard. edu/ExpressDB. We
recommend standards for data reporting, suggest research into improving
comparability of microarray data through quantifying and standardizing control
condition RNA populations, and also suggest research into the calibration of
different RNA assays. We introduce a model for a database that integrates
different kinds of functional genomics data, Biomolecule Interaction, Growth and
Expression Database (BIGED). |
Which CYP gene polymorphism is a well-known predictor of efavirenz disposition? | Cytochrome P450 (CYP) CYP2B6 G516T (rs3745274) is a well-known predictor of efavirenz disposition. | Background and Objectives: Severe, recalcitrant cases of pediatric psoriasis or
atopic dermatitis may necessitate treatment with biological agents; however,
this may be difficult due to lack of treatment options and standardized
treatment guidelines. This review evaluates the biological treatment options
available, including off-label uses, and provides a basic therapeutic guideline
for pediatric psoriasis and atopic dermatitis. Materials and Methods: A PubMed
review of biological treatments for pediatric psoriasis and atopic dermatitis
with information regarding age, efficacy, dosing, contra-indications, adverse
events, and off-label treatments. Results: Currently there are three European
Medicines Agency (EMA)-approved biological treatment options for pediatric
psoriasis: etanercept, ustekinumab, and adalimumab. While dupilumab was recently
Food and Drug Administration (FDA)- and EMA-approved for adult atopic
dermatitis, it is still not yet approved for pediatric atopic dermatitis.
Conclusions: Given the high morbidity associated with pediatric atopic
dermatitis and psoriasis, there is a need for more treatment options. Further
research and post-marketing registries are needed to extend the use of biologics
into pediatric patients. Author information:
(1)Clinical Pharmacology Department, Instituto Teófilo Herdo, Universidad
Autónoma de Madrid (UAM) Instituto de Investigación Sanitaria La Princesa (IP),
Hospital Universitario de La Princesa, Madrid, Spain.
(2)UICEC Hospital Universitario de La Princesa, Plataforma SCReN (Spanish
Clinical Research Network), Instituto de Investigación Sanitaria La Princesa
(IP), Madrid, Spain.
(3)Clinical Pharmacology Department, Instituto Teófilo Herdo, Universidad
Autónoma de Madrid (UAM) Instituto de Investigación Sanitaria La Princesa (IP),
Hospital Universitario de La Princesa, Madrid, Spain.
[email protected].
(4)UICEC Hospital Universitario de La Princesa, Plataforma SCReN (Spanish
Clinical Research Network), Instituto de Investigación Sanitaria La Princesa
(IP), Madrid, Spain. [email protected].
(5)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y
Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.
[email protected].
(6)Pharmacology Department, Facultad de Medicina, Universidad Autónoma de
Madrid, Madrid, Spain. [email protected]. |
What is the role of the IRE1a-XBP1 pathway? | The IRE1a-XBP1 pathway is a conserved adaptive mediator of the unfolded protein response, playing an important role in the regulation of cell differentiation. | BACKGROUND: The IRE1a-XBP1 pathway is a conserved adaptive mediator of the
unfolded protein response. The pathway is indispensable for the development of
secretory cells by facilitating protein folding and enhancing secretory
capacity. In the immune system, it is known to function in dendritic cells,
plasma cells, and eosinophil development and differentiation, while its role in
T helper cell is unexplored. Here, we investigated the role of the IRE1a-XBP1
pathway in regulating activation and differentiation of type-2 T helper cell
(Th2), a major T helper cell type involved in allergy, asthma, helminth
infection, pregcy, and tumor immunosuppression.
METHODS: We perturbed the IRE1a-XBP1 pathway and interrogated its role in Th2
cell differentiation. We performed genome-wide transcriptomic analysis of
differential gene expression to reveal IRE1a-XBP1 pathway-regulated genes and
predict their biological role. To identify direct target genes of XBP1 and
define XBP1's regulatory network, we performed XBP1 ChIPmentation (ChIP-seq). We
validated our predictions by flow cytometry, ELISA, and qPCR. We also used a
fluorescent ubiquitin cell cycle indicator mouse to demonstrate the role of XBP1
in the cell cycle.
RESULTS: We show that Th2 lymphocytes induce the IRE1a-XBP1 pathway during in
vitro and in vivo activation. Genome-wide transcriptomic analysis of
differential gene expression by perturbing the IRE1a-XBP1 pathway reveals
XBP1-controlled genes and biological pathways. Performing XBP1 ChIPmentation
(ChIP-seq) and integrating with transcriptomic data, we identify XBP1-controlled
direct target genes and its transcriptional regulatory network. We observed that
the IRE1a-XBP1 pathway controls cytokine secretion and the expression of two Th2
signature cytokines, IL13 and IL5. We also discovered that the IRE1a-XBP1
pathway facilitates activation-dependent Th2 cell proliferation by facilitating
cell cycle progression through S and G2/M phase.
CONCLUSIONS: We confirm and detail the critical role of the IRE1a-XBP1 pathway
during Th2 lymphocyte activation in regulating cytokine expression, secretion,
and cell proliferation. Our high-quality genome-wide XBP1 ChIP and gene
expression data provide a rich resource for investigating XBP1-regulated genes.
We provide a browsable online database available at http://data.teichlab.org . |
Can Panitumumab cause trichomegaly? | Yes. Panitumumab is EGFR inhibitor that is associated with eyelash trichomegaly. | Eyelash trichomegaly is an uncommon drug-associated sequelae experienced during
treatment with epidermal growth factor receptor (EGFR) inhibitors. Elongation of
the eyelashes induced by these agents has predomitly been observed in
oncology patients with either colorectal or lung cancer. It is most frequently
associated with cetuximab and erlotinib; however, it has also been described in
individuals treated with gefitinib or panitumumab. We describe
cetuximab-associated eyelash trichomegaly in a woman with metastatic rectal
carcinoma. We review the clinical presentation, adverse effects, and management
of EGFR inhibitor-related eyelash trichomegaly. The long eyelashes are not a
drug-limiting adverse effect and some patients consider the change to be
cosmetically enhancing. Trimming the lashes with scissors can usually ameliorate
local symptoms. The eyelashes often return to their original length at variable
time periods after EGFR inhibitor therapy is discontinued. A wide spectrum of skin toxicities has been described in patients receiving
epidermal growth factor receptor (EGFR), inhibitors, including papulopustular
rash, xerosis and fissures, pruritus, mucositis, paronychia, and hair
changes.Trichomegaly of the eyelashes is a rare adverse effect of EGFR inhibitor
therapy and is characterized by a paradoxical overgrowth of eyelashes. We
present 3 cases of trichomegaly occurred during EGFR inhibitor therapy. |
List proteins that are contained in atherosclerotic plaques? | extracellular matrix proteins
biglycan
Lumican
Apolipoprotein A-I | Atherosclerosis is a chronic inflammatory disease with complex pathobiology and
one of the most common causes of cardiovascular events. The process is
characterized by complex vascular remodeling processes that require the actions
of numerous proteins. The composition of atherosclerotic plaque is increasingly
recognized as a major factor governing the occurrence of cardiovascular or
neurological symptoms. To gain deeper insights into the composition of
atherosclerotic plaques, we created quantitative proteome profiles of advanced
plaque tissues of six male patients undergoing carotid endarterectomy for stroke
prevention. Using a quantitative, data-independent proteome approach, we
identified 4181 proteins with an average protein coverage of 45%. An analysis of
the quantitative composition of the tissue revealed key players of vascular
remodeling processes. Moreover, compared with proximal arterial tissue, 20
proteins in mature plaques were enriched, whereas 52 proteins were found in
lower quantities. Among the proteins with increased abundance were prominent
extracellular matrix proteins such as biglycan and lumican, whereas cytoskeletal
markers for contractile smooth muscle cells (SMCs) were decreased. Taken
together, this study provides the most comprehensive quantitative assessment of
mature human plaque tissue to date, which indicates a central role of SMCs in
the structure of advanced atherosclerotic plaques. To evaluate the presence of serum protein biomarkers associated with the early
phases of formation of carotid atherosclerotic plaques, label-free quantitative
proteomics analyses were made for serum samples collected as part of The
Cardiovascular Risk in Young Finns Study. Samples from subjects who had an
asymptomatic carotid artery plaque detected by ultrasound examination (N = 43,
Age = 30-45 years) were compared with plaque free controls (N = 43) (matched for
age, sex, body weight and systolic blood pressure). Seven proteins (p < 0.05)
that have been previously linked with atherosclerotic phenotypes were
differentially abundant. Fibulin 1 proteoform C (FBLN1C),
Beta-ala-his-dipeptidase (CNDP1), Cadherin-13 (CDH13), Gelsolin (GSN) and 72 kDa
type IV collagenase (MMP2) were less abundant in cases, whereas Apolipoproteins
C-III (APOC3) and apolipoprotein E (APOE) were more abundant. Using machine
learning analysis, a biomarker panel of FBLN1C, APOE and CDH13 was identified,
which classified cases from controls with an area under receiver-operating
characteristic curve (AUROC) value of 0.79. Furthermore, using selected reaction
monitoring mass spectrometry (SRM-MS) the decreased abundance of FBLN1C was
verified. In relation to previous associations of FBLN1C with atherosclerotic
lesions, the observation could reflect its involvement in the initiation of the
plaque formation, or represent a particular risk phenotype. Arterial foam cells are central players of atherogenesis. Cholesterol acceptors,
apolipoprotein A-I (apoA-I) and high-density lipoprotein (HDL), take up
cholesterol and phospholipids effluxed from foam cells into the circulation. Due
to the high abundance of cholesterol in foam cells, most previous studies
focused on apoA-I/HDL-mediated free cholesterol (FC) transport. However, recent
lipidomics of human atherosclerotic plaques also identified that oxidized
sterols (oxysterols) and non-sterol lipid species accumulate as atherogenesis
progresses. While it is known that these lipids regulate expression of
pro-inflammatory genes linked to plaque instability, how cholesterol acceptors
impact the foam cell lipidome, particularly oxysterols and non-sterol lipids,
remains unexplored. Using lipidomics analyses, we found cholesterol acceptors
remodel foam cell lipidomes. Lipid subclass analyses revealed various
oxysterols, sphingomyelins, and ceramides, species uniquely enriched in human
plaques were significantly reduced by cholesterol acceptors, especially by
apoA-I. These results indicate that the function of lipid-poor apoA-I is not
limited to the efflux of cholesterol and phospholipids but suggest that apoA-I
serves as a major regulator of the foam cell lipidome and might play an
important role in reducing multiple lipid species involved in the pathogenesis
of atherosclerosis. Cardiovascular disease (CVD) is the most common cause of death in industrialized
countries. One underlying cause is atherosclerosis, which is a systemic disease
characterized by plaques of retained lipids, inflammatory cells, apoptotic
cells, calcium and extracellular matrix (ECM) proteins in the arterial wall. The
biologic composition of an atherosclerotic plaque determines whether the plaque
is more or less vulnerable, that is prone to rupture or erosion. Here, the ECM
and tissue repair play an important role in plaque stability, vulnerability and
progression. This review will focus on ECM remodelling in atherosclerotic
plaques, with focus on how ECM biomarkers might predict plaque vulnerability and
outcome. |
List characteristics of Developmental and Epileptic Encephalopathies (DEEs). | yes, developmental and epileptic encephalopathies (dees) are a group of severe, early onset epilepsies characterized by refractory seizures, developmental delay or regression associated with ongoing epileptic activity, and generally poor prognosis. | Author information:
(1)Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC
H3T1C5, Canada.
(2)Department of Pediatrics, Division of Genetic Medicine, University of
Washington, Seattle, WA 98195, USA.
(3)Centre Hospitalier de l'Université de Montréal Research Center, Montreal, QC
H2X 0A9, Canada; Department of Neurosciences, Université de Montréal, Montreal,
QC H3T1J4, Canada.
(4)Montreal Neurological Institute, Department of Neurology and Neurosurgery,
McGill University, Montreal, QC H3A2B4, Canada.
(5)McGill University and Genome Quebec Innovation Center, Montreal, QC H3A 1A4,
Canada; Department of Human Genetics, McGill University, Montreal, QC H3A 1B1,
Canada.
(6)Centre Hospitalier de l'Université de Montréal Research Center, Montreal, QC
H2X 0A9, Canada; Center for Pediatric Genomic Medicine, Children's Mercy Kansas
City, Kansas City, MO 64108, USA; Department of Pathology and Laboratory
Medicine, Children's Mercy Kansas City, Kansas City, MO 64108, USA.
(7)Centre Hospitalier de l'Université de Montréal Research Center, Montreal, QC
H2X 0A9, Canada.
(8)GeneDx, Gaithersburg, MD 20877, USA.
(9)Department of Molecular and Human Genetics, Baylor College of Medicine,
Houston, TX 77030, USA.
(10)Department of Molecular and Human Genetics, Baylor College of Medicine,
Houston, TX 77030, USA; Baylor Miraca Genetics Laboratories, Baylor College of
Medicine, Houston, TX 77021, USA.
(11)Program in Genetics and Genome Biology, Division of Neurology, Department of
Pediatrics, Hospital for Sick Children and University of Toronto, Toronto, ON
M5G 0A4, Canada.
(12)Division of Neurology, Epilepsy Genetics Program, Krembil Neuroscience
Centre, Toronto Western Hospital, University of Toronto, Toronto, ON M5G 2C4,
Canada.
(13)Epilepsy Research Centre, Department of Medicine, University of Melbourne,
Austin Health, Heidelberg, VIC 3084, Australia.
(14)Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK.
(15)MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine,
University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK.
(16)Clinical Genetics Service, University Hospitals Bristol NHS Foundation
Trust, St. Michael's Hospital, St. Michael's Hill, Bristol BS2 8DT, UK.
(17)North West Thames Regional Genetics Service, London North West Healthcare
NHS Trust, Northwick Park Hospital, Watford Road, Harrow HA1 3UJ, UK.
(18)Oxford Centre for Genomic Medicine, ACE building Nuffield Orthopaedic
Centre, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 7HE, UK.
(19)Manchester Centre for Genomic Medicine, St. Mary's Hospital, Central
Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health
Science Centre, Manchester M13 9WL, UK.
(20)Institute of Medical Genetics, University Hospital of Wales, Heath Park,
Cardiff CF14 4XW, UK.
(21)MRC Centre for Neuropsychiatric Genetics & Genomics, Hadyn Ellis Building,
Cathays, Cardiff University, Cardiff CF24 4HQ, UK.
(22)West of Scotland Regional Genetics Service, Queen Elizabeth University
Hospital, Glasgow G51 4TF, UK.
(23)North East Thames Regional Genetics Service, Great Ormond Street Hospital
for Children, London WC1N 3JH, UK.
(24)Yorkshire Regional Genetics Service, Leeds Teaching Hospitals NHS Trust,
Department of Clinical Genetics, Chapel Allerton Hospital, Chapeltown Road,
Leeds LS7 4SA, UK.
(25)Department of Pediatrics, Section of Medical Genetics, SUNY Upstate Medical
University, Syracuse, NY 13210, USA.
(26)University of Groningen, University Medical Center Groningen, Department of
Genetics, 9700 RB Groningen, the Netherlands.
(27)University of South Dakota Sanford School of Medicine, Sioux Falls, SD
57117, USA.
(28)Augustana-Sanford Genetic Counseling Graduate Program, Sioux Falls, SD
57197, USA.
(29)Departments of Medicine and Pediatrics, Columbia University Medical Center,
New York, NY 10032, USA.
(30)Baptist Hospital, Miami, FL 33176 USA.
(31)Joe DiMaggio Children's Hospital, Hollywood, FL 33021, USA.
(32)Division of Genetics and Genomic Medicine, Department of Pediatrics,
Washington University School of Medicine, St. Louis, MO 63110, USA.
(33)Department of Human Genetics, Donders Centre for Brain, Cognition and
Behavior, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands.
(34)Centre de Génétique des Anomalies du Développement, Centre Hospitalier
Universitaire de Dijon, 21000 Dijon, France; Équipe INSERM 1231, Génétique des
Anomalies du Développement, Université de Bourgogne, 21000 Dijon, France.
(35)Genetics Department, Assistance Publique - Hôpitaux de Paris, Robert-Debré
University Hospital, 75000 Paris, France.
(36)Department of Clinical Genetics, United Laboratories, Tartu University
Hospital and Institute of Clinical Medicine, University of Tartu, Tartu 51014,
Estonia.
(37)Division of Genetics and Genomics and Division of Newborn Medicine,
Department of Medicine, Boston Children's Hospital, Harvard Medical School,
Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142,
USA.
(38)Nationwide Children's Hospital and Ohio State University, Department of
Pediatrics, Division of Neurology, Columbus, OH 43205, USA.
(39)Division of Neurology, Children's Hospital of Eastern Ontario, Ottawa, ON
K1H 8L1, Canada.
(40)University of Tasmania, Royal Hobart Hospital, Department of Paediatrics,
Hobart, TAS 7000, Australia.
(41)School of Medicine, University of Tasmania, Hobart, TAS 7000, Australia.
(42)Population Health and Immunity Division, Walter and Eliza Hall Institute of
Medical Research, Parkville, VIC 3052, Australia; Epilepsy Research Centre,
Department of Medicine, University of Melbourne, Austin Health, Heidelberg, VIC
3084, Australia.
(43)Children's Hospital at Westmead Clinical School, University of Sydney,
Westmead, NSW 2145, Australia.
(44)Department of Molecular and Human Genetics, Baylor College of Medicine,
Houston, TX 77030, USA; Texas Children's Hospital, Houston, TX 77030, USA.
(45)Dipartimento di Pediatria e di Neuropsichiatria Infantile, Università La
Sapienza, 00185 Rome, Italy.
(46)Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di
Sanità, 00161 Rome, Italy.
(47)Genetics and Rare Diseases Research Division, Bambino Gesù Children's
Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, 00165 Rome,
Italy.
(48)Metabolic Neurogenetic Clinic and Pediatric Movement Disorders Clinic,
Wolfson Medical Center, Holon 5822012, Israel.
(49)University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
(50)University of British Columbia, BC Children's Hospital, Vancouver, BC V6H
3N1, Canada.
(51)Department of Medical Genetics, University of British Columbia, Vancouver,
BC V6H 3N1, Canada.
(52)Département de Génétique, Centre de Référence des Déficiences
Intellectuelles de Causes Rares, Groupe de Recherche Clinique "Déficiences
Intellectuelles et Autisme," Université Pierre et Marie Curie, Hôpital de la
Pitié-Salpêtrière, Paris 75013, France; Sorbonne Universités, Université Pierre
et Marie Curie (Université Paris 06), UMRS 1127, INSERM U 1127, CNRS UMR 7225,
Institut du Cerveau et de la Moelle Épinière, Paris 75013, France.
(53)Assistance Publique - Hôpitaux de Paris, Hôpital d'Enfants Armand Trousseau,
Service de Neuropédiatrie, Paris 75012, France.
(54)Université Paris Diderot, Sorbonne Paris Cité, INSERM UMR 1141, Paris 75019,
France; Assistance Publique - Hôpitaux de Paris, Hôpital Robert Debré, Service
de Neurologie Pédiatrique, Paris 75019, France.
(55)HudsonAlpha Institute for Biotechnology, 601 Genome Way, Huntsville, AL
35806, USA.
(56)Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
35294, USA.
(57)Department of Pediatrics, University of Arkansas for Medical Sciences,
Little Rock, AR 72205, USA; Arkansas Children's Research Institute, Little Rock,
AR 72205, USA.
(58)Texas Children's Hospital and Baylor College of Medicine, Houston, TX 77030,
USA.
(59)Centre Hospitalier Rouyn-Noranda, Rouyn-Noranda, QC J9X 2B2, Canada.
(60)Division of Neurology, Centre Hospitalier Universitaire de Québec, Quebec,
QC G1V 4G2, Canada.
(61)Department of Pediatrics, Centre Hospitalier Universitaire de Sherbrooke,
Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
(62)Department of Pediatrics, McGill University, Montreal, QC H3A 1A4, Canada;
Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A
1A4, Canada.
(63)Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal,
QC H3T1C5, Canada; Department of Neurosciences, Université de Montréal,
Montreal, QC H3T1J4, Canada; Department of Pediatrics, Université de Montréal,
Montreal, QC H3T1C5, Canada.
(64)Department of Neurosciences, Université de Montréal, Montreal, QC H3T1J4,
Canada; Department of Pediatrics, Université de Montréal, Montreal, QC H3T1C5,
Canada.
(65)Department of Pediatrics and Child Health, University of Otago, Wellington
9016, New Zealand.
(66)Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal,
QC H3T1C5, Canada; Department of Pediatrics, Université de Montréal, Montreal,
QC H3T1C5, Canada.
(67)Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal,
QC H3T1C5, Canada; Department of Neurosciences, Université de Montréal,
Montreal, QC H3T1J4, Canada.
(68)Centre Hospitalier de l'Université de Montréal Research Center, Montreal, QC
H2X 0A9, Canada; Department of Human Genetics, McGill University, Montreal, QC
H3A 1B1, Canada; Département des Sciences Fondamentales, Université du Québec à
Chicoutimi, Chicoutimi, QC G7H 2B1, Canada.
(69)Division of Clinical and Metabolic Genetics, Department of Pediatrics,
University of Toronto, The Hospital for Sick Children, Toronto, ON M5G 1X8,
Canada.
(70)Division of Neurology, BC Children's Hospital, Vancouver, BC V6H 3N1,
Canada.
(71)Epilepsy Research Centre, Department of Medicine, University of Melbourne,
Austin Health, Heidelberg, VIC 3084, Australia; Department of Pediatrics,
University of Melbourne Royal Children's Hospital, Parkville, VIC 3052,
Australia; Florey Institute of Neuroscience and Mental Health, Melbourne, VIC
3084, Australia.
(72)Program in Genetics and Genome Biology, Division of Neurology, Department of
Pediatrics, Hospital for Sick Children and University of Toronto, Toronto, ON
M5G 0A4, Canada; Division of Child Neurology, Department of Pediatrics,
University of Texas Southwestern, Dallas, TX 75390, USA. Electronic address:
[email protected].
(73)Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal,
QC H3T1C5, Canada; Department of Neurosciences, Université de Montréal,
Montreal, QC H3T1J4, Canada; Department of Pediatrics, Université de Montréal,
Montreal, QC H3T1C5, Canada. Electronic address:
[email protected]. Author information:
(1)UF Innovation en diagnostic genomique des maladies rares, CHU Dijon
Bourgogne, Dijon, France. [email protected].
(2)INSERM UMR1231 GAD, F-21000, Dijon, France.
[email protected].
(3)Universite Claude Bernard Lyon I, CHU de Lyon, Lyon, France.
(4)Service de Radiologie, Hopital-Femme-Mère-Enfant, Hospices Civils de Lyon,
Lyon, France.
(5)INSERM UMR1231 GAD, F-21000, Dijon, France.
(6)Departement de Genetique, Hopital Pitie-Salpetriere, Paris, France.
(7)Division of Genetics and Metabolic Phoenix Children's Hospital, Phoenix,
Arizona, USA.
(8)Inserm U 1127, CNRS UMR 7225, Sorbonne Universites, UPMC Univ Paris 06 UMR S
1127, Institut du Cerveau et de la Moelle epinière, ICM, Paris, France.
(9)Reference Center for Adult Neurometabolic Diseases, Pitie-Salpêtrière
University Hospital, Paris, France.
(10)Department of Genetics, University Medical Center, Utrecht, The Netherlands.
(11)Centre de Reference maladies rares « Anomalies du Developpement et syndrome
malformatifs » de l'Est, Centre de Genetique, Hopital d'Enfants, FHU TRANSLAD,
CHU Dijon Bourgogne, Dijon, France.
(12)UF Innovation en diagnostic genomique des maladies rares, CHU Dijon
Bourgogne, Dijon, France.
(13)Department of Child Neurology, Brain Center Rudolf Magnus, University
Medical Center, Utrecht, The Netherlands.
(14)Department of Pediatrics, Division of Medical Genetics, Cedars-Sinai Medical
Center and Harbor-UCLA Medical Center, Los Angeles, California, USA.
(15)Division of Pediatric Neurology, Department of Pediatrics, Harbor-UCLA
Medical Center, Los Angeles, California, USA.
(16)GeneDx, Gaithersburg, Maryland, USA.
(17)Genomed Ltd., Moscow, Russia.
(18)Veltischev Research and Clinical Institute for Pediatrics of the Pirogov
Russian National Research Medical University, Moscow, Russia.
(19)Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical
Center, Torrance, California, USA.
(20)Departments of Human Genetics and Psychiatry, David Geffen School of
Medicine at UCLA, Los Angeles, California, USA.
(21)Institute of Medical Genetics, University of Zurich, Schlieren, Zurich,
Switzerland.
(22)Division of Pediatric Neurology, Children's Hospital, Lucerne, Switzerland.
(23)Department of Neurosciences and Pediatrics UCSD/Rady Children's Hospital San
Diego, Rady Children's Institute for Genomic Medicine, San Diego, California,
USA.
(24)UF Innovation en diagnostic genomique des maladies rares, CHU Dijon
Bourgogne, Dijon, France. [email protected].
(25)INSERM UMR1231 GAD, F-21000, Dijon, France.
[email protected]. Author information:
(1)Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA
19104, USA.
(2)Department of Neurology and Epileptology, Hertie Institute for Clinical Brain
Research, University of Tübingen, 72076 Tübingen, Germany.
(3)Department of Physiology & Pharmacology, Hotchkiss Brain Institute and
Alberta Children's Hospital Research Institute, University of Calgary, Calgary,
AB T2N 1N4, Canada.
(4)Division of Genetic Medicine, University of Washington, Seattle, WA 98195,
USA.
(5)APHP, Département de Génétique, Centre de Référence Déficiences
Intellectuelles de Causes Rares, Groupe Hospitalier Pitié Salpêtrière et GHUEP
Hôpital Trousseau; Sorbonne Université, GRC "Déficience Intellectuelle et
Autisme," 75013 Paris, France.
(6)Sorbonne Université, GRC n°19, Pathologies Congénitales du
Cervelet-LeucoDystrophies, Département de Génétique et Embryologie Médicale,
AP-HP, Hôpital d'Enfants Armand Trousseau, Centre de Référence des Déficits
Intellectuels de Causes Rares, 75012 Paris, France.
(7)Sorbonne Université, GRC n°19, Pathologies Congénitales du
Cervelet-LeucoDystrophies, Service de Neuropédiatrie, AP-HP, Hôpital d'Enfants
Armand Trousseau; Centre de Référence des Déficits Intellectuels de Causes
Rares; Inserm U 1141, 75012 Paris, France.
(8)Department of Clinical Genetics, Odense University Hospital, 5000 Odense,
Denmark; H.C. Andersen Children's Hospital, Odense University Hospital, 5000
Odense, Denmark.
(9)Department of Clinical Genetics, Odense University Hospital, 5000 Odense,
Denmark.
(10)Department of Human Genetics, Radboud University Medical Center, 6525
Nijmegen, the Netherlands.
(11)Stichting Epilepsie Instellingen Nederland, 8025 Zwolle, the Netherlands.
(12)Department of Neurology, Academic Center for Epileptology, Kempenhaeghe and
Maastricht UMC, 5591 Heeze, the Netherlands.
(13)Barrow Neurological Institute, Phoenix Children's Hospital, Departments of
Child Health, Genetics, Neurology, and Cellular & Molecular Medicine, University
of Arizona College of Medicine, Phoenix, AZ 85013, USA.
(14)Division of Genetics and Metabolism, Phoenix Children's Hospital, Phoenix,
AZ 85016, USA.
(15)Yale School of Medicine, New Haven, CT 06510, USA.
(16)Department of Pediatric Neurology, Penteli Children's Hospital, 152 36
Athens, Greece.
(17)Institute of Biomedical Science and Children's Hospital Fudan University,
201102 Shanghai, China.
(18)Perinatal Center, Sahlgrenska Academy, Gothenburg University, 413 46
Gothenburg, Sweden; Hospital of Zhengzhou University, 450001 Zhengzhou, China.
(19)Epilepsy Research Centre, Department of Medicine, The University of
Melbourne, Austin Health, Heidelberg, VIC 3084, Australia.
(20)Department of Health Science Research, Mayo Clinic, Rochester, MN 55905,
USA.
(21)Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN
55905, USA.
(22)Department of Health Science Research, Mayo Clinic, Rochester, MN 55905,
USA; Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA.
(23)Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA;
Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA.
(24)University of Illinois Chicago College of Medicine, University of Illinois
College of Medicine at Peoria, Peoria, IL 61605, USA.
(25)Adult Genetics Unit, Royal Adelaide Hospital, and School of Medicine,
University of Adelaide, Adelaide, SA 5000, Australia.
(26)Department of Neurology, Women's and Children's Hospital, University of
Adelaide, North Adelaide, SA 5006, Australia.
(27)Center for Human Disease Modeling, Duke University Medical Center, Durham,
NC 27701, USA.
(28)Adelaide Medical School, Robinson Research Institute, University of
Adelaide, North Adelaide, SA 5006, Australia.
(29)CeGaT, 72076 Tübingen, Germany.
(30)Department of Human Genetics, University of Tübingen, 72076 Tübingen,
Germany.
(31)Division of Genetics and Genomics and Department of Neurology, Boston
Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard
Medical School, Boston, MA 02215, USA.
(32)Northeast Regional Epilepsy Group, Hackensack University Medical Center,
Hackensack, NJ 07601, USA.
(33)University of Louisville, Louisville, KY 40292, USA.
(34)Children's Hospital of Wisconsin, Milwaukee, WI 53226, USA.
(35)Division of Medical Genetics, Department of Pediatrics, University of Utah,
Salt Lake City, UT 84113, USA.
(36)Division of Pediatric Neurology, Departments of Pediatrics and Neurology,
University of Utah, Salt Lake City, UT 84113, USA.
(37)ARUP Laboratories, Salt Lake City, UT 84108, USA.
(38)Division of Metabolic Disorders CHOC Children's Hospital, Orange, CA 92868,
USA.
(39)Division of Metabolic Disorders CHOC Children's Hospital, Orange, CA 92868,
USA; Department of Pediatrics, University of California-Irvine School of
Medicine, Irvine, CA 92617, USA.
(40)Centre for Brain Research and School of Biological Sciences, The University
of Auckland, Auckland 1142, New Zealand.
(41)Department of Neurology, Starship Children's Health, Auckland 1023, New
Zealand.
(42)Department of Paediatrics and Child Health, University of Otago Wellington,
Wellington South 6242, New Zealand.
(43)Department of Clinical Genetics, Karolinska University Hospital, 171 76
Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska
Institutet, 171 77 Stockholm, Sweden.
(44)Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 77
Stockholm, Sweden.
(45)Department of Neuroscience, Azienda Ospedaliero-Universitaria Meyer,
University of Florence, 50139 Florence, Italy.
(46)Greenwood Genetic Center, Greenwood, SC 29646, USA.
(47)Medical Genetics, Sanford Health, Bemidji, MN 56601, USA.
(48)Medical Genetics, Sanford Health, Fargo, ND 58102, USA.
(49)Epilepsy Genetics Program, Department of Neurology, Boston Children's
Hospital, Boston, MA 02115, USA; Department of Neurology, Harvard Medical
School, Boston, MA 02215, USA.
(50)Epilepsy Genetics Program, Department of Neurology, Boston Children's
Hospital, Boston, MA 02115, USA.
(51)Department of Neurology, Harvard Medical School, Boston, MA 02215, USA;
Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA.
(52)Department of Haematology, University of Cambridge, NHS Blood and Transplant
Centre, Cambridge CB2 0QQ, UK; NIHR BioResource - Rare Diseases, Cambridge
University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus,
Cambridge CB2 0QQ, UK.
(53)Clinical Genetics, Royal Devon and Exeter NHS Foundation Trust, Exeter EX2
5DW, UK.
(54)Department of Neurology, Royal Devon and Exeter NHS Foundation Trust, Exeter
EX2 5DW, UK.
(55)NIHR BioResource - Rare Diseases, Cambridge University Hospitals NHS
Foundation Trust, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Department
of Medical Genetics, Cambridge Institute for Medical Research, University of
Cambridge, Cambridge CB2 0XY, UK.
(56)Yorkshire Regional Genetics Service, Chapel Allerton Hospital Leeds Teaching
Hospitals NHS Trust, Leeds LS7 4SA, UK.
(57)Division of Evolution and Genomic Sciences, School of Biological Sciences,
Faculty of Biology, Medicine and Health, The University of Manchester,
Manchester M13 9PL, UK; Manchester Centre for Genomic Medicine, St. Mary's
Hospital, Manchester University Foundation NHS Trust, Health Innovation,
Manchester M13 9WL, UK.
(58)Department of Paediatric Neurology, Royal Manchester Children's Hospital,
Manchester University Foundation NHS Trust, Health Innovation, Manchester M13
9WL, UK.
(59)Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester
University Foundation NHS Trust, Health Innovation, Manchester M13 9WL, UK.
(60)Wellcome Sanger Institute, Cambridge CB10 1SA, UK.
(61)Epilepsy Research Centre, Department of Medicine, The University of
Melbourne, Austin Health, Heidelberg, VIC 3084, Australia; The Florey Institute
and Murdoch Children's Research Institute, Parkville, VIC 3052, Australia;
Department of Paediatrics, The University of Melbourne, Royal Children's
Hospital, Parkville, VIC 3052, Australia; Department of Neurology, Royal
Children's Hospital, Parkville, VIC 3052, Australia.
(62)Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA
19104, USA; Department of Neuropediatrics, Christian-Albrechts-University of
Kiel, 24105 Kiel, Germany.
(63)Division of Genetic Medicine, University of Washington, Seattle, WA 98195,
USA. Electronic address: [email protected]. De Novo Pathogenic Variants in CACNA1E Cause Developmental and Epileptic
Encephalopathy With Contractures, Macrocephaly, and Dyskinesias Helbig KL,
Lauerer RJ, Bahr JC, et al. Am J Hum Genet. 2019;104(3):562. Developmental and
epileptic encephalopathies (DEEs) are severe neurodevelopmental disorders often
beginning in infancy or early childhood that are characterized by intractable
seizures, abundant epileptiform activity on electroencephalogram (EEG), and
developmental impairment or regression. CACNA1E is highly expressed in the
central nervous system and encodes the α1-subunit of the voltage-gated CaV2.3
channel, which conducts high-voltage-activated R-type calcium currents that
initiate synaptic transmission. Using next-generation sequencing techniques, we
identified de novo CACNA1E variants in 30 individuals with DEE, characterized by
refractory infantile-onset seizures, severe hypotonia, and profound
developmental impairment, often with congenital contractures, macrocephaly,
hyperkinetic movement disorders, and early death. Most of the 14, partially
recurring, variants cluster within the cytoplasmic ends of all 4 S6 segments,
which form the presumed CaV2.3 channel activation gate. Functional analysis of
several S6 variants revealed consistent gain-of-function effects comprising
facilitated voltage-dependent activation and slowed inactivation. Another
variant located in the domain II S4-S5 linker results in facilitated activation
and increased current density. Five participants achieved seizure freedom on the
antiepileptic drug topiramate, which blocks R-type calcium channels. We
establish pathogenic variants in CACNA1E as a cause of DEEs and suggest
facilitated R-type calcium currents as a disease mechanism for human epilepsy
and developmental disorders. OBJECTIVE: Developmental epileptic encephalopathies (DEEs) are genetically
heterogeneous severe childhood-onset epilepsies with developmental delay or
cognitive deficits. In this study, we explored the pathogenic mechanisms of
DEE-associated de novo mutations in the CACNA1A gene.
METHODS: We studied the functional impact of four de novo DEE-associated CACNA1A
mutations, including the previously described p.A713T variant and three novel
variants (p.V1396M, p.G230V, and p.I1357S). Mutant cDNAs were expressed in
HEK293 cells, and whole-cell voltage-clamp recordings were conducted to test the
impacts on CaV 2.1 channel function. Channel localization and structure were
assessed with immunofluorescence microscopy and three-dimensional (3D) modeling.
RESULTS: We find that the G230V and I1357S mutations result in loss-of-function
effects with reduced whole-cell current densities and decreased channel
expression at the cell membrane. By contrast, the A713T and V1396M variants
resulted in gain-of-function effects with increased whole-cell currents and
facilitated current activation (hyperpolarized shift). The A713T variant also
resulted in slower current decay. 3D modeling predicts conformational changes
favoring channel opening for A713T and V1396M.
SIGNIFICANCE: Our findings suggest that both gain-of-function and
loss-of-function CACNA1A mutations are associated with similarly severe DEEs and
that functional validation is required to clarify the underlying molecular
mechanisms and to guide therapies. Developmental and Epileptic encephalopathies (DEE) describe heterogeneous
epilepsy syndromes, characterized by early-onset, refractory seizures and
developmental delay (DD). Several DEE associated genes have been reported. With
increased access to whole exome sequencing (WES), new candidate genes are being
identified although there are fewer large cohort papers describing the clinical
phenotype in such patients. We describe 6 unreported individuals and provide
updated information on an additional previously reported individual with
heterozygous de novo missense variants in YWHAG. We describe a syndromal
phenotype, report 5 novel, and a recurrent p.Arg132Cys YWHAG variant and compare
developmental trajectory and treatment strategies in this cohort. We provide
further evidence of causality in YWHAG variants. WES was performed in five
patients via Deciphering Developmental Disorders Study and the remaining two
were identified via Genematcher and AnnEX databases. De novo variants identified
from exome data were validated using Sanger sequencing. Seven out of seven
patients in the cohort have de novo, heterozygous missense variants in YWHAG
including 2/7 patients with a recurrent c.394C > T, p.Arg132Cys variant; 1/7 has
a second, pathogenic variant in STAG1. Characteristic features included:
early-onset seizures, predomitly generalized tonic-clonic and absence type
(7/7) with good response to standard anti-epileptic medications; moderate DD;
Intellectual Disability (ID) (5/7) and Autism Spectrum Disorder (3/7). De novo
YWHAG missense variants cause EE, characterized by early-onset epilepsy, ID and
DD, supporting the hypothesis that YWHAG loss-of-function causes a neurological
phenotype. Although the exact mechanism of disease resulting from alterations in
YWHAG is not fully known, it is possible that haploinsufficiency of YWHAG in
developing cerebral cortex may lead to abnormal neuronal migration resulting in
DEE. BACKGROUND: Early-onset developmental and epileptic encephalopathy (DEE) is
characterized by repeated seizures beginning within 3 months of birth and severe
interictal epileptiform discharge, including burst suppression. This study
assessed the utility of targeted gene panel sequencing in the genetic diagnosis
of this disease.
MATERIALS AND METHODS: Targeted gene panel sequencing was performed in 150 early
infantile-onset DEE patients (≤3 months of age), and we extensively reviewed
their clinical characteristics, including therapeutic efficacy, according to
genotype.
RESULTS: Of the early infantile-onset DEE patients, 70 were neonatal-onset DEE
and the other 80 patients began experiencing seizures from 1 to 3 months after
birth. There were 11 different pathogenic or likely pathogenic variants among
34.7% (52/150) of patients with early infantile-onset DEE, in whom KCNQ2,
STXBP1, CDKL5, and SCN1A were the major pathogenic variants. Among the
neonatal-onset DEE patients, pathological genes were identified in 42.9%
(30/70), indicating a significantly higher diagnostic yield than in 27.5%
(22/80) of patients who experienced seizure onset 1 to 3 months after birth
(p = 0.048). Among the neonatal-onset DEE group, variants in KCNQ2, STXBP1, and
CDKL5 were detected at high frequencies, accounting for 66.7% (20/30) of the
pathogenic or likely pathogenic variants found in this study.
CONCLUSION: Targeted gene panel sequencing demonstrated a high yield of
pathogenic variants in the diagnosis of early-onset epileptic encephalopathy,
especially in those with neonatal-onset DEE. Early diagnosis of early-onset
epileptic encephalopathy may improve the prognosis of patients by earlier
selection of appropriate treatment based on pathogenic variant. Developmental and epileptic encephalopathies (DEEs) are a group of severe, early
onset epilepsies characterized by refractory seizures, developmental delay or
regression associated with ongoing epileptic activity, and generally poor
prognosis. DEE is genetically and phenotypically heterogeneous, and there is a
plethora of genetic testing options to investigate the rapidly growing list of
epilepsy genes. However, more than 50% of patients with DEE remain without a
genetic diagnosis despite state-of-the-art genetic testing. In this review, we
discuss the major advances in epilepsy genomics that have surfaced in recent
years. The goal of this review is to reach a larger audience and build a better
understanding of pathogenesis and genetic testing options in DEE. Developmental and epileptic encephalopathies (DEE) are a heterogeneous group of
disorders characterized by epilepsy with comorbid intellectual disability.
Recently, two de novo heterozygous mutations in the gene encoding TRPM3, a
calcium permeable ion channel, were identified as the cause of DEE in eight
probands, but the functional consequences of the mutations remained elusive.
Here we demonstrate that both mutations (V990M and P1090Q) have distinct effects
on TRPM3 gating, including increased basal activity, higher sensitivity to
stimulation by the endogenous neurosteroid pregnenolone sulfate (PS) and heat,
and altered response to ligand modulation. Most strikingly, the V990M mutation
affected the gating of the non-canonical pore of TRPM3, resulting in large
inward cation currents via the voltage sensor domain in response to PS
stimulation. Taken together, these data indicate that the two DEE mutations in
TRPM3 result in a profound gain of channel function, which may lie at the basis
of epileptic activity and neurodevelopmental symptoms in the patients. AIM: Developmental and epileptic encephalopathies (DEEs) are a group of
devastating disorders caused by epileptic activity, resulting in deterioration
in developmental, cognitive, and motor functions. The number of genes identified
as being responsible for DEEs has been increasing rapidly. However, despite a
comprehensive molecular analysis, a molecular diagnosis can only be established
in 50% of cases. The aim of this project is to use whole exome sequencing (WES)
to determine the molecular etiology of DEEs in undiagnosed patients with a
pedigree suggestive of an autosomal recessive single gene disease.
METHODS: Three DEE families, having either consanguineous parents of an affected
individual and/or having more than one affected offspring, were enrolled in the
project. Prior to this project, the families had been evaluated using a
next-generation sequencing panel including 16 DEE genes in a previous study;
however, no molecular diagnosis could be established. In five cases from the
three selected DEEs families in our study, the genetic etiology was investigated
using WES.
RESULTS: All patients in the study group had infantile onset epileptic seizures;
however, semiologies varied. All patients presented with severe developmental
delay. WES revealed biallelic disease causing mutations in DENDD5A, GRN, and
TBCD genes in family 1, family 2, and family 3, respectively. In each family,
the identified variants associated with the disease were segregated. Reverse
phenotyping supported the molecular analysis.
CONCLUSION: This study provided a valuable contribution to the
genotype-phenotype relationship by determining rare epilepsy syndromes in
undiagnosed patients previously. WES is a useful diagnostic alternative,
particularly in consanguineous families. Developmental and epileptic encephalopathies (DEEs) are the spectrum of severe
epilepsies characterized by early-onset, refractory seizures occurring in the
context of developmental regression or plateauing. Early infantile epileptic
encephalopathy (EIEE) is one of the earliest forms of DEE, manifesting as
frequent epileptic spasms and characteristic electroencephalogram findings in
early infancy. In recent years, next-generation sequencing approaches have
identified a number of monogenic determits underlying DEE. In the case of
EIEE, 85 genes have been registered in Online Mendelian Inheritance in Man as
causative genes. Model organisms are indispensable tools for understanding the
in vivo roles of the newly identified causative genes. In this review, we first
present an overview of epilepsy and its genetic etiology, especially focusing on
EIEE and then briefly summarize epilepsy research using animal and
patient-derived induced pluripotent stem cell (iPSC) models. The Drosophila
model, which is characterized by easy gene manipulation, a short generation
time, low cost and fewer ethical restrictions when designing experiments, is
optimal for understanding the genetics of DEE. We therefore highlight studies
with Drosophila models for EIEE and discuss the future development of their
practical use. OBJECTIVE: SCN2A-associated developmental and epileptic encephalopathies (DEEs)
present with seizures, developmental impairments, and often both. We sought to
characterize the level and pattern of development in children with SCN2A
variants, and to address the sensitivity of the Vineland Adaptive Behavior
Scales (VABS) in measuring changes over time in children with SCN2A-DEEs.
METHODS: Clinical histories for participants with pathogenic SCN2A variants in
the Simons SearchLight project were analyzed for descriptive purposes. VABS
scores obtained at study entry and yearly thereafter were analyzed for floor and
ceiling effects, change with age, and association with epilepsy through use of
regression and longitudinal regression methods.
RESULTS: Sixty-four participants (50 with epilepsy, 30 [47%] female, median age
49 months, interquartile range [IQR] 28 to 101) were included. Histories of
birth complications (N = 34, 54%), neonatal neurological signs (N = 45, 74%),
and other neurological symptoms (N = 31, 48%) were common and similar in
epilepsy and nonepilepsy subgroups. Mean standardized VABS scores (Composite
53.5; Motor, 55.8, Communication, 54.1, Socialization, 59.4, and Daily living
skills, 55.1) reflected performance ~3 standard deviations below the normative
test average. In longitudinal regression analyses, standardized scores decreased
between 1.3 and 2.8 points per year, suggesting regression of abilities. Raw
score analyses, however, revealed several subdomains with substantial floor
effects (eg, community use); other raw scores increased with increasing age.
Participants with epilepsy scored 0.6 to 1 SD lower than those without epilepsy
(all P's < .05).
SIGNIFICANCE: The VABS, as standardly administered, has shortcomings for
addressing growth or regression in individuals with SCN2A-DEEs. Some subdomain
raw scores reflected substantial floor effects. Raw scores increased so slowly
over time that standardized scores declined. Alternative measures sensitive to
incremental meaningful change are required if outcomes such as adaptive behavior
are to be primary outcomes in short-term clinical trials. |
Which methods exist for efficient calculation of Elementary flux modes (EFMs) in genome-scale metabolic networks (GSMNs)? | EFM-Ta is a novel algorithm that uses a linear programming-based tree search and efficiently enumerates a subset of EFMs in genome-scale metabolic networks (GSMNs). The stand-alone software TreeEFM is implemented in C++ and interacts with the open-source linear solver COIN-OR Linear program Solver (CLP). | MOTIVATION: Elementary flux modes (EFMs) analysis constitutes a fundamental tool
in systems biology. However, the efficient calculation of EFMs in genome-scale
metabolic networks (GSMNs) is still a challenge. We present a novel algorithm
that uses a linear programming-based tree search and efficiently enumerates a
subset of EFMs in GSMNs.
RESULTS: Our approach is compared with the EFMEvolver approach, demonstrating a
significant improvement in computation time. We also validate the usefulness of
our new approach by studying the acetate overflow metabolism in the Escherichia
coli bacteria. To do so, we computed 1 million EFMs for each energetic amino
acid and then analysed the relevance of each energetic amino acid based on
gene/protein expression data and the obtained EFMs. We found good agreement
between previous experiments and the conclusions reached using EFMs. Finally, we
also analysed the performance of our approach when applied to large GSMNs.
AVAILABILITY AND IMPLEMENTATION: The stand-alone software TreeEFM is implemented
in C++ and interacts with the open-source linear solver COIN-OR Linear program
Solver (CLP). |
What is the mechanism of action of vosoritide? | Vosoritide is a biologic analogue of C-type natriuretic peptide, a potent stimulator of endochondral ossification. | TransCon CNP is a C-type natriuretic peptide (CNP-38) conjugated via a cleavable
linker to a polyethylene glycol carrier molecule, designed to provide sustained
systemic CNP levels upon weekly subcutaneous administration. TransCon CNP is in
clinical development for the treatment of comorbidities associated with
achondroplasia. In both mice and cynomolgus monkeys, sustained exposure to CNP
via TransCon CNP was more efficacious in stimulating bone growth than
intermittent CNP exposure. TransCon CNP was well tolerated with no adverse
cardiovascular effects observed at exposure levels exceeding the expected
clinical therapeutic exposure. At equivalent dose levels, reductions in blood
pressure and/or an increase in heart rate were seen following single
subcutaneous injections of the unconjugated CNP-38 molecule or a daily CNP-39
molecule (same amino acid sequence as Vosoritide, USAN:INN). The half-life of
the daily CNP-39 molecule in cynomolgus monkey was estimated to be 20 minutes,
compared with 90 hours for CNP-38, released from TransCon CNP. C max for the
CNP-39 molecule (20 µg/kg) was approximately 100-fold higher, compared with the
peak CNP level associated with administration of 100 µg/kg CNP as TransCon CNP.
Furthermore, CNP exposure for the daily CNP-39 molecule was only evident for up
to 2 hours postdose (lower limit of quantification 37 pmol/l), whereas TransCon
CNP gave rise to systemic exposure to CNP-38 for at least 7 days postdose. The
prolonged CNP exposure and associated hemodynamically safe peak serum
concentrations associated with TransCon CNP administration are suggested to
improve efficacy, compared with short-lived CNP molecules, due to better
therapeutic drug coverage and decreased risk of hypotension. SIGNIFICANCE
STATEMENT: The hormone C-type natriuretic peptide (CNP) is in clinical
development for the treatment of comorbidities associated with achondroplasia,
the most common form of human dwarfism. The TransCon Technology was used to
design TransCon CNP, a prodrug that slowly releases active CNP in the body over
several days. Preclinical data show great promise for TransCon CNP to be an
effective and well-tolerated drug that provides sustained levels of CNP in a
convenient once-weekly dose, while avoiding high systemic CNP bolus
concentrations that can induce cardiovascular side effects. BACKGROUND: Achondroplasia is a genetic disorder that inhibits endochondral
ossification, resulting in disproportionate short stature and clinically
significant medical complications. Vosoritide is a biologic analogue of C-type
natriuretic peptide, a potent stimulator of endochondral ossification.
METHODS: In a multinational, phase 2, dose-finding study and extension study, we
evaluated the safety and side-effect profile of vosoritide in children (5 to 14
years of age) with achondroplasia. A total of 35 children were enrolled in four
sequential cohorts to receive vosoritide at a once-daily subcutaneous dose of
2.5 μg per kilogram of body weight (8 patients in cohort 1), 7.5 μg per kilogram
(8 patients in cohort 2), 15.0 μg per kilogram (10 patients in cohort 3), or
30.0 μg per kilogram (9 patients in cohort 4). After 6 months, the dose in
cohort 1 was increased to 7.5 μg per kilogram and then to 15.0 μg per kilogram,
and in cohort 2, the dose was increased to 15.0 μg per kilogram; the patients in
cohorts 3 and 4 continued to receive their initial doses. At the time of data
cutoff, the 24-month dose-finding study had been completed, and 30 patients had
been enrolled in an ongoing long-term extension study; the median duration of
follow-up across both studies was 42 months.
RESULTS: During the treatment periods in the dose-finding and extension studies,
adverse events occurred in 35 of 35 patients (100%), and serious adverse events
occurred in 4 of 35 patients (11%). Therapy was discontinued in 6 patients (in 1
because of an adverse event). During the first 6 months of treatment, a
dose-dependent increase in the annualized growth velocity was observed with
vosoritide up to a dose of 15.0 μg per kilogram, and a sustained increase in the
annualized growth velocity was observed at doses of 15.0 and 30.0 μg per
kilogram for up to 42 months.
CONCLUSIONS: In children with achondroplasia, once-daily subcutaneous
administration of vosoritide was associated with a side-effect profile that
appeared generally mild. Treatment resulted in a sustained increase in the
annualized growth velocity for up to 42 months. (Funded by BioMarin
Pharmaceutical; ClinicalTrials.gov numbers, NCT01603095, NCT02055157, and
NCT02724228.). |
What is the "flight-or-fight response"? | This is also known as " flight-or- fight response" and is defined as an individual's response to a stimulus such as stress, that includes loss of sleep, anxiety, shortness of breath, muscle and joint pain. | The brain is the key organ of the response to stress because it determines what
is threatening and, therefore, potentially stressful, as well as the
physiological and behavioral responses which can be either adaptive or damaging.
Stress involves two-way communication between the brain and the cardiovascular,
immune, and other systems via neural and endocrine mechanisms. Beyond the
"flight-or-fight" response to acute stress, there are events in daily life that
produce a type of chronic stress and lead over time to wear and tear on the body
("allostatic load"). Yet, hormones associated with stress protect the body in
the short-run and promote adaptation ("allostasis"). The brain is a target of
stress, and the hippocampus was the first brain region, besides the
hypothalamus, to be recognized as a target of glucocorticoids. Stress and stress
hormones produce both adaptive and maladaptive effects on this brain region
throughout the life course. Early life events influence life-long patterns of
emotionality and stress responsiveness and alter the rate of brain and body
aging. The hippocampus, amygdala, and prefrontal cortex undergo stress-induced
structural remodeling, which alters behavioral and physiological responses. As
an adjunct to pharmaceutical therapy, social and behavioral interventions such
as regular physical activity and social support reduce the chronic stress burden
and benefit brain and body health and resilience. The fear, flight or fight response serves as the fundamental physiological basis
for examining an organism's awareness of its environment under an impending
predator attack. Although it is not known whether invertebrates possess an
autonomic nervous system identical to that of vertebrates, evidence shows
invertebrates have a sympathetic-like response to regulate the internal
environment and ready the organism to act behaviorally to a given stimuli.
Furthermore, this physiological response can be feasibly measured and it acts as
a biological index for the animal's internal state. Measurements of the
physiological response can be directly related to internal and external
stressors through changes in the central nervous system controlled coordination
of the cardio-vascular and respiratory systems. More specifically, monitoring
heart and ventilation rates provide quantifiable measures of the stress response
not always behaviorally observed. Crayfish are good model organisms for heart
and ventilatory rate measurements due to the feasibility of recording, as well
as the rich history known of the morphology of the crayfish, dating back to
Huxley in 1888, and the well-studied typical behaviors. In the complex microcosm of a cell, information security and its faithful
transmission are critical for maintaining internal stability. To achieve a
coordinated response of all its parts to any stimulus the cell must protect the
information received from potentially confounding signals. Physical segregation
of the information transmission chain ensures that only the entities able to
perform the encoded task have access to the relevant information. The cAMP
intracellular signaling pathway is an important system for signal transmission
responsible for the ancestral 'flight or fight' response and involved in the
control of critical functions including frequency and strength of heart
contraction, energy metabolism and gene transcription. It is becoming
increasingly apparent that the cAMP signaling pathway uses compartmentalization
as a strategy for coordinating the large number of key cellular functions under
its control. Spatial confinement allows the formation of cAMP signaling "hot
spots" at discrete subcellular domains in response to specific stimuli, bringing
the information in proximity to the relevant effectors and their recipients,
thus achieving specificity of action. In this report we discuss how the
different constituents of the cAMP pathway are targeted and participate in the
formation of cAMP compartmentalized signaling events. We illustrate a few
examples of localized cAMP signaling, with a particular focus on the nucleus,
the sarcoplasmic reticulum and the mitochondria. Finally, we discuss the
therapeutic potential of interventions designed to perturb specific cAMP
cascades locally. Although the function of the autonomic nervous system (ANS) in mediating the
flight-or-fight response was recognized decades ago, the crucial role of
peripheral innervation in regulating cell behavior and response to the
microenvironment has only recently emerged. In the hematopoietic system, the ANS
regulates stem cell niche homeostasis and regeneration and fine-tunes the
inflammatory response. Additionally, emerging data suggest that cancer cells
take advantage of innervating neural circuitry to promote their progression.
These new discoveries outline the need to redesign therapeutic strategies to
target this underappreciated stromal constituent. Here, we review the importance
of neural signaling in hematopoietic homeostasis, inflammation, and cancer. |
How is Burke-Fahn-Marsden Dystonia scale used? | Burck-Fahn-Marsden Dystonia scale (BBS) is a simple, reliable, and valid measure of disease severity in patients with dystonia. | Object. Deep brain stimulation (DBS) of the globus pallidus internus (GPi) is a
promising new procedure for the treatment of dystonia. The authors present their
technical approach for placement of electrodes into the GPi in awake patients
with dystonia, including the methodology used for electrophysiological mapping
of the GPi in the dystonic state, clinical outcomes and complications, and the
location of electrodes associated with optimal benefit. Methods. Twenty-three
adult and pediatric patients who had various forms of dystonia were included in
this study. Baseline neurological status and improvement in motor function
resulting from DBS were measured using the Burke-Fahn-Marsden Dystonia Rating
Scale (BFMDRS). Implantation of the DBS lead was performed using magnetic
resoce (MR) imaging-based stereotaxy, single-cell microelectrode recording,
and intraoperative test stimulation to determine thresholds for
stimulation-induced adverse effects. Electrode locations were measured on
computationally reformatted postoperative MR images according to a prospective
protocol. Conclusions. Physiologically guided implantation of DBS electrodes in
patients with dystonia is technically feasible in the awake state in most cases,
with low morbidity rates. Spontaneous discharge rates of GPi neurons in dystonia
are similar to those of globus pallidus externus neurons, such that the two
nuclei must be distinguished by neuronal discharge patterns rather than by
rates. Active electrode locations associated with robust improvement (> 50%
decrease in BFMDRS score) were located near the intercommissural plane, at a
mean distance of 3.7 mm from the pallidocapsular border. Patients with
juvenile-onset primary dystonia and those with the tardive form benefited
greatly from this procedure, whereas benefits for most secondary dystonias and
the adult-onset craniocervical form of this disorder were more modest. Seven children between 2 and 15 years of age with cerebral palsy and upper
extremity dystonia were enrolled in an open-label, dose-escalation pilot
clinical trial of botulinum toxin type B (Myobloc), injected into the biceps and
brachioradialis muscles of I or both arms. The primary outcome measure was the
change in maximum speed of hand movement during attempted forward reaching.
Escalating doses of 12.5, 25, and 50 U/kg per muscle were injected at each of 3
visits. Reaching speed improved in response to injection, and dystonia scores on
the Burke-Fahn-Marsden dystonia scale, the Unified Dystonia Rating Scale, and
the Unified Parkinson's Disease Rating Scale improved. There was not a
dose-related effect on efficacy. There were no serious adverse events. Two
children reported transient weakness. These results support the use of botulinum
toxin type B as a safe and effective treatment for upper extremity dystonia in
children with cerebral palsy. Larger controlled trials are needed to confirm
these results. Neurodegeneration with brain iron accumulation encompasses a heterogeneous group
of rare neurodegenerative disorders that are characterized by iron accumulation
in the brain. Severe generalized dystonia is frequently a prominent symptom and
can be very disabling, causing gait impairment, difficulty with speech and
swallowing, pain and respiratory distress. Several case reports and one case
series have been published concerning therapeutic outcome of pallidal deep brain
stimulation in dystonia caused by neurodegeneration with brain iron
degeneration, reporting mostly favourable outcomes. However, with case studies,
there may be a reporting bias towards favourable outcome. Thus, we undertook
this multi-centre retrospective study to gather worldwide experiences with
bilateral pallidal deep brain stimulation in patients with neurodegeneration
with brain iron accumulation. A total of 16 centres contributed 23 patients with
confirmed neurodegeneration with brain iron accumulation and bilateral pallidal
deep brain stimulation. Patient details including gender, age at onset, age at
operation, genetic status, magnetic resoce imaging status, history and
clinical findings were requested. Data on severity of dystonia (Burke Fahn
Marsden Dystonia Rating Scale-Motor Scale, Barry Albright Dystonia Scale),
disability (Burke Fahn Marsden Dystonia Rating Scale-Disability Scale), quality
of life (subjective global rating from 1 to 10 obtained retrospectively from
patient and caregiver) as well as data on supportive therapy, concurrent
pharmacotherapy, stimulation settings, adverse events and side effects were
collected. Data were collected once preoperatively and at 2-6 and 9-15 months
postoperatively. The primary outcome measure was change in severity of dystonia.
The mean improvement in severity of dystonia was 28.5% at 2-6 months and 25.7%
at 9-15 months. At 9-15 months postoperatively, 66.7% of patients showed an
improvement of 20% or more in severity of dystonia, and 31.3% showed an
improvement of 20% or more in disability. Global quality of life ratings showed
a median improvement of 83.3% at 9-15 months. Severity of dystonia
preoperatively and disease duration predicted improvement in severity of
dystonia at 2-6 months; this failed to reach significance at 9-15 months. The
study confirms that dystonia in neurodegeneration with brain iron accumulation
improves with bilateral pallidal deep brain stimulation, although this
improvement is not as great as the benefit reported in patients with primary
generalized dystonias or some other secondary dystonias. The patients with more
severe dystonia seem to benefit more. A well-controlled, multi-centre
prospective study is necessary to enable evidence-based therapeutic decisions
and better predict therapeutic outcomes. Primary Meige syndrome is an idiopathic movement disorder that manifests as
craniofacial and often cervical dystonias. Deep brain stimulation (DBS) of the
globus pallidus internus (GPi) has emerged as a powerful surgical option in the
treatment of primary generalized or segmental dystonia. However, the experience
with GPi-DBS in Meige syndrome is limited. We followed 5 patients with disabling
Meige syndrome treated by bilateral GPi-DBS for 49 ± 43.7 (mean ± SD) months.
All patients were assessed before surgery and at the last follow-up after
surgery using the Burke-Fahn-Marsden Dystonia Rating Scale (BFMDRS) which
includes both the movement and disability scales. Bilateral GPi-DBS produced a
sustained and long-lasting improvement in dystonia symptoms associated with
Meige syndrome. At the last follow-up, the mean scores of BFMDRS movement and
disability scales improved significantly by 84 ± 6.8% (range, 75-94%) and
89 ± 8.1% (range, 80-100%), respectively. Bilateral pallidal stimulation is a
beneficial therapeutic option for long-term relief of the disabling dystonia
symptoms in Meige syndrome. BACKGROUND: The preoperative evaluation in dystonia aims at characterizing the
severity and topography of motor symptoms in patients, who have previously been
selected for deep brain stimulation (DBS).
METHODS: The literature search was performed using PubMed, CINAHL, and the
Cochrane Collaborative databases.
RESULTS: Commonly used scales for clinical assessment are the Burke-Fahn-Marsden
dystonia rating scale for generalized dystonia and the Toronto Western Spasmodic
Torticollis Scale for cervical dystonia. Motor assessment is completed by
quality of life and functional scales, such as the Short-Form Health Survey
(SF-36) or the Parkinson's Disease Questionnaire 39. Validated rating scales for
cranial or upper limb dystonia are lacking.
DISCUSSION: In common clinical practice, these outcome measures can be
administered in an open-label fashion because double blind assessment is only
required for ascertaining new treatment indications or research purposes. The
same measures are to be used postoperatively to revaluate outcome after DBS.
Brain MRI is required to confirm diagnosis and assess structural abnormalities.
Other imaging techniques, particularly functional imaging, are used for research
purposes. Secondary dystonia encompasses a heterogeneous group with different etiologies.
Cerebral palsy is the most common cause. Pharmacological treatment is often
unsatisfactory. There are only limited data on the therapeutic outcomes of deep
brain stimulation in dyskinetic cerebral palsy. The published literature
regarding deep brain stimulation and secondary dystonia was reviewed in a
meta-analysis to reevaluate the effect on cerebral palsy. The Burke-Fahn-Marsden
Dystonia Rating Scale movement score was chosen as the primary outcome measure.
Outcome over time was evaluated and summarized by mixed-model repeated-measures
analysis, paired Student t test, and Pearson's correlation coefficient. Twenty
articles comprising 68 patients with cerebral palsy undergoing deep brain
stimulation assessed by the Burke-Fahn-Marsden Dystonia Rating Scale were
identified. Most articles were case reports reflecting great variability in the
score and duration of follow-up. The mean Burke-Fahn-Marsden Dystonia Rating
Scale movement score was 64.94 ± 25.40 preoperatively and dropped to 50.5 ±
26.77 postoperatively, with a mean improvement of 23.6% (P < .001) at a median
follow-up of 12 months. The mean Burke-Fahn-Marsden Dystonia Rating Scale
disability score was 18.54 ± 6.15 preoperatively and 16.83 ± 6.42
postoperatively, with a mean improvement of 9.2% (P < .001). There was a
significant negative correlation between severity of dystonia and clinical
outcome (P < .05). Deep brain stimulation can be an effective treatment option
for dyskinetic cerebral palsy. In view of the heterogeneous data, a prospective
study with a large cohort of patients in a standardized setting with a
multidisciplinary approach would be helpful in further evaluating the role of
deep brain stimulation in cerebral palsy. © 2013 Movement Disorder Society. BACKGROUND/AIMS: Reports of outcomes in treating dystonia secondary to stroke
with deep brain stimulation (DBS) are limited. We report our experience with 3
patients, all with infarcts involving the striatum, who developed hemidystonia
and were treated with unilateral globus pallidus interna DBS.
METHODS: Case series describing characteristics and outcomes based on the
Burke-Fahn-Marsden Dystonia Rating Scale (BFMDRS) scores before and after DBS at
3, 6 and at least 12 months.
RESULTS: All patients reported subjective improvements after surgery. At 1 year
or more after surgery, none of the 3 patients displayed a measureable
improvement in the BFMDRS movement score.
CONCLUSION: Our findings are consistent with previous reports of limited
benefits from pallidal DBS in secondary dystonia. Future work should focus on
predictive factors for DBS outcomes and the development of more sensitive
assessment tools specifically for secondary dystonias as well as the exploration
of alternative brain targets for stimulation. PURPOSE: To evaluate the functional goal-directed outcomes of Deep Brain
Stimulation (DBS) in childhood dystonia according to aetiology and to explore
relationship with a traditional impairment-based measure.
METHOD: This is a prospective case series study involving thirty children with
dystonia with a 1-year follow-up post-DBS. The Canadian Occupational Performance
Measure (COPM) and Burke-Fahn-Marsden Dystonia Rating Scale (BFMDRS) were used
as primary outcome measures. Results were analysed based on aetiology in 3
groups: 1. primary/primary plus dystonia; 2. secondary dystonia-cerebral palsy
(CP); 3. secondary dystonia-non-CP group. Correlation between functional outcome
using COPM and dystonia improvement as captured by BFMDRS was measured.
RESULTS: All groups demonstrated significant improvement in individualised goal
attainment, measured with the COPM, at 1-year post-DBS. The secondary
dystonia-CP group also achieved significant improvement at 6 months for
performance and satisfaction scores. In the majority of secondary dystonias, the
BFMDRS failed to demonstrate significant improvement. A linear correlation
between change in BFMDRS and COPM scores was observed when the entire cohort was
analysed.
INTERPRETATION/CONCLUSIONS: DBS improved functional performance, independently
of the dystonic phenotype. Improvements in individualized COPM functional goal
areas were seen in the absence of significant changes in BFMDRS scores,
highlighting the relative insensitivity of impairment scales in this patient
group. AIM: Hyperkinetic movement disorders (HMDs) can be assessed using
impairment-based scales or functional classifications. The Burke-Fahn-Marsden
Dystonia Rating Scale-movement (BFM-M) evaluates dystonia impairment, but may
not reflect functional ability. The Gross Motor Function Classification System
(GMFCS), Manual Ability Classification System (MACS), and Communication Function
Classification System (CFCS) are widely used in the literature on cerebral palsy
to classify functional ability, but not in childhood movement disorders. We
explore the concordance of these three functional scales in a large sample of
paediatric HMDs and the impact of dystonia severity on these scales.
METHOD: Children with HMDs (n=161; median age 10y 3mo, range 2y 6mo-21y) were
assessed using the BFM-M, GMFCS, MACS, and CFCS from 2007 to 2013. This
cross-sectional study contrasts the information provided by these scales.
RESULTS: All four scales were strongly associated (all Spearman's rank
correlation coefficient rs >0.72, p<0.001), with worse dystonia severity
implying worse function. Secondary dystonias had worse dystonia and less
function than primary dystonias (p<0.001). A longer proportion of life lived
with dystonia is associated with more severe dystonia (rs =0.42, p<0.001).
INTERPRETATION: The BFM-M is strongly linked with the GMFCS, MACS, and CFCS,
irrespective of aetiology. Each scale offers interrelated but complementary
information and is applicable to all aetiologies. Movement disorders including
cerebral palsy can be effectively evaluated using these scales. Pallidal deep brain stimulation (DBS) is an established treatment for patients
with severe isolated dystonia. However, clinical evidence for the long-term use
of DBS in children is limited and controlled trials have not yet been conducted.
Here, we provide the long-term results of up to 13 years of pallidal DBS in
eight pediatric patients with generalized idiopathic or hereditary isolated
dystonia (five males, mean age at surgery 12.5 ± 3.5 years), as assessed by
retrospective video rating. Video rating was performed at three time points:
pre-operative, 1-year short-term follow-up (1y-FU) and long-term last FU (LT-FU,
up to 13 years). Symptom severity and disability were assessed using the
Burke-Fahn-Marsden Dystonia Rating Scale (BFMDRS). Disability scores were
obtained from clinical charts and during the last FU. The mean improvement in
BFMDRS motor score was 54.4 ± 8.9 % at 1y-FU and 42.9 ± 11.6 % at LT-FU; the
disability scores improved by 59.8 ± 10.3 and 63.3 ± 7.8 %, respectively.
Electrode dislocation was noted in one patient and implantable pulse generator
dislocation in another, both requiring surgical intervention; no further serious
adverse events occurred. Our study presents the first blinded video rating
assessment of the short- and long-term effects of pallidal DBS in children with
idiopathic or hereditary isolated dystonia. Results confirm that pallidal DBS is
a safe and efficacious long-term treatment in children, with overall motor
improvement similar to that described in controlled trials in adults. BACKGROUND AND PURPOSE: Huntington's disease (HD) is an autosomal domit,
neurodegenerative movement disorder, typically characterized by chorea. Dystonia
is also recognized as part of the HD motor phenotype, although little work
detailing its prevalence, distribution, severity and impact on functional
capacity has been published to date.
METHODS: Patients (>18 years of age) were recruited from the Cardiff (UK) HD
clinic, each undergoing a standardized videotaped clinical examination and
series of functional assessment questionnaires (Unified Huntington's Disease
Rating Scale, Burke-Fahn-Marsden Dystonia Rating Scale and modified version of
the Toronto Western Spasmodic Torticollis Rating Scale). The presence and
severity of dystonia were scored by four independent neurologists using the
Burke-Fahn-Marsden Dystonia Rating Scale and Unified Huntington's Disease Rating
Scale. Statistical analysis included Fisher's exact test, Wilcoxon test, anova
and calculation of correlation coefficients where appropriate.
RESULTS: Forty-eight patients [91% (48/53)] demonstrated evidence of dystonia,
with the highest prevalence in the left upper limb (n = 44, 83%), right upper
limb most severely affected and eyes least affected. Statistically significant
positive correlations (P < 0.05) were observed between dystonia severity and
increasing HD disease stage and motor disease duration. Deterioration in
functional capacity also correlated with increasing dystonia severity. No
significant relationship was observed with age at motor symptom onset or CAG
repeat length.
CONCLUSIONS: We report a high prevalence of dystonia in adult patients with HD,
with worsening dystonia severity with increasing HD disease stage and motor
disease duration. The recognition and management of dystonic symptoms in routine
clinical practice will aid overall symptomatic treatment and functional
improvement. BACKGROUND: The Burke-Fahn-Marsden Dystonia Rating Scale is a universally
applied instrument for the quantitative assessment of dystonia in both children
and adults. However, immature movements by healthy young children may also show
"dystonic characteristics" as a consequence of physiologically incomplete brain
maturation. This could implicate that Burke-Fahn-Marsden scale scores are
confounded by pediatric age.
OBJECTIVE: In healthy young children, we aimed to determine whether
physiologically immature movements and postures can induce an age-related effect
on Burke-Fahn-Marsden movement and disability scale scores.
METHODS: Nine assessors specializied in movement disorders (3 adult
neurologists, 3 pediatric neurologists, and 3 MD/PhD students) independently
scored the Burke-Fahn-Marsden movement scale in 52 healthy children (4-16 years
of age; 2 boys and 2 girls per year of age). Independent of that, parents scored
their children's functional motor development according to the
Burke-Fahn-Marsden disability scale in another 52 healthy children (4-16 years
of age; 2 boys and 2 girls per year of age). By regression analysis, we
determined the association between Burke-Fahn-Marsden movement and disability
scales outcomes and pediatric age.
RESULTS: In healthy children, assessment of physiologically immature motor
performances by the Burke-Fahn-Marsden movement and disability scales showed an
association between the outcomes of both scales and age (until 16 years and 12
years of age, β = -0.72 and β = -0.60, for Burke-Fahn-Marsden movement and
disability scale, respectively [both P < 0.001]).
CONCLUSIONS: The Burke-Fahn-Marsden movement and disability scales are
influenced by the age of the child. For accurate interpretation of longitudinal
Burke-Fahn-Marsden Dystonia Rating Scale scores in young dystonic children,
consideration of pediatric age-relatedness appears advisory. OBJECTIVES: The pallidothalamic tract connects the globus pallidus internus with
the ventroanterior and ventrolateral parts of the thalamus. Lesioning or
stimulation of the pallidothalamic tract has ameliorating effects on dyskinesia
and dystonia in patients with Parkinson disease. However, the effect of the
procedure on dystonia due to other etiologies has not been reported.
METHODS: We retrospectively analyzed patients with dystonia who underwent
unilateral pallidothalamic tractotomy between July 2017 and October 2018 at
Tokyo Women's Medical University Hospital. The Burke-Fahn-Marsden Dystonia
Rating Scale-Movement Scale was used to evaluate the severity of dystonia at
three time points (before surgery, 3 months postoperatively, and the last
available follow-up). Adverse events were also evaluated.
RESULTS: Eleven patients underwent unilateral pallidothalamic tractotomy,
including 5 with generalized dystonia, 4 with segmental dystonia, and 2 with
focal (cervical) dystonia. All patients had undergone unilateral pallidotomy
before contralateral pallidothalamic tractotomy. The mean interval between the
previous surgery (pallidotomy) and pallidothalamic tractotomy was 9.5 ± 3.1
months. The mean follow-up period was 11.5 ± 4.2 months. The Burke-Fahn-Marsden
Dystonia Rating Scale-Movement Scale scores at 3 months after pallidothalamic
tractotomy (5.8 ± 8.4) and at the last available follow-up (5.6 ± 8.3, P <
0.001) were significantly improved compared with that before pallidothalamic
tractotomy (21.8 ± 16.3). The most common adverse event was reduced voice volume
(6 patients), which was mild and did not interfere with the patient's daily
activities.
CONCLUSIONS: This study suggests that pallidothalamic tractotomy can be an
alternative treatment target for dystonia. A larger and longer prospective study
is needed to elucidate the safety and efficacy of pallidothalamic tractotomy for
dystonia. OBJECTIVE: To evaluate the short-term and long-term clinical effectiveness and
safety of subthalamic nucleus deep brain stimulation (STN-DBS) for medically
intractable pediatric isolated dystonia.
METHODS: Using a longitudinal retrospective design, we assessed the clinical
outcomes of nine patients who underwent STN-DBS for treatment-refractory
pediatric isolated dystonia one decade ago (mean age at surgery: 15.9 ± 4.5
years). The primary clinical outcome used was assessed by retrospective video
analyses of patients' dystonia symptoms using the Burke-Fahn-Marsden Dystonia
Rating Scale (BFMDRS). Clinical assessments were performed at baseline, 1-year
follow-up (1-yr FU), and 10-year follow-up (10-yr FU). Adverse side effects,
including surgery-related, device-related, and stimulation-related effects, were
also documented.
RESULTS: After STN-DBS surgery, the mean improvement in the BFMDRS motor score
was 77.1 ± 26.6% at 1-yr FU and 90.4 ± 10.4% at 10-yr FU. Similarly, the mean
BFMDRS disability score was improved by 69.5 ± 13.6% at 1-yr FU and by 86.5 ±
13.9% at 10-yr FU. The clinical improvements gained at 10-yr FU were
significantly larger than those observed at 1-yr FU. Negative correlations were
found between the duration of disease to age at surgery ratio (DD/AS) and the
improvements in the BFMDRS motor score and total score at 1-yr FU and 10-yr FU.
CONCLUSION: To our knowledge, this study provides the first clinical evidence
for the short- and long-term effectiveness and safety of STN-DBS for pediatric
isolated dystonia. Additionally, putative evidence is provided that earlier
STN-DBS intervention in patients with refractory pediatric isolated dystonia may
improve short- and long-term clinical outcomes. AIM: To establish the prevalence of dystonic pain in children and their response
to deep brain stimulation (DBS).
METHOD: Dystonic pain was assessed in a cohort of 140 children, 71 males and 69
females, median age 11 years 11 months (range 3y-19y 1mo), undergoing DBS in our
centre over a period of 10 years. The cohort was divided into aetiological
dystonia groups: 1a, inherited; 1b, heredodegenerative; 2, acquired; and 3,
idiopathic. Motor responses were measured with the Burke-Fahn-Marsden Dystonia
Rating Scale (BFMDRS).
RESULTS: Dystonic pain was identified in 63 (45%) patients, 38% of whom had a
diagnosis of cerebral palsy (CP). Dystonic pain improved in 90% of children and
in all aetiological subgroups 1 year after DBS, while the BFMDRS motor score
improved in 70%. Statistically significant improvement (p<0.01) was noted for
the whole cohort on the Numerical Pain Rating Scale (n=27), Paediatric Pain
Profile (n=17), and Caregivers Priorities and Child Health Index of Life with
Disabilities questionnaire (n=48). There was reduction of pain severity,
frequency, and analgesia requirement. Findings were similar for the whole cohort
and aetiological subgroups other than the inherited heredodegenerative group
where the improvement did not reach statistical significance.
INTERPRETATION: Dystonic pain is frequent in children with dystonia, including
those with CP, who undergo DBS; this can be an important, realizable goal of
surgery irrespective of aetiology. We encourage the use of multimodal approach
in pain research to reduce the risk of bias. BACKGROUND: Although an increasing number of trials are reported on the
treatment of generalized or segmental isolated dystonia, the minimal clinically
important difference thresholds for the most frequently reported outcome
measures are still undetermined.
OBJECTIVES: To estimate the minimal clinically important difference for the
Burke-Fahn-Marsden Dystonia Rating Scale and the 36-Item Short-Form Health
Survey in generalized or segmental dystonia.
METHODS: A total of 898 paired examinations of 198 consecutive patients, aged
>18 years, with idiopathic and inherited (torsin family 1 member A positive)
segmental and generalized isolated dystonia were analyzed. To calculate the
minimal clinically important difference thresholds, both anchor- and
distribution-based methods were used simultaneously.
RESULTS: Any improvement >16.6% or worsening larger than 21.5% on the
Burke-Fahn-Marsden Dystonia Rating Scale indicates a minimal, yet clinically
relevant, change. Threshold values for the Burke-Fahn-Marsden Dystonia
Disability Scale were 0.5 points for both decline and improvement. Cut-off
scores for the Physical Component Summary, the Mental Component Summary, and the
Global (Total or Overall) Score of the 36-Item Short-Form Health Survey were 5.5
and 5.5, 6.5 and 7.5, and 7.5 and 8.5 points for clinically meaningful
improvement and deterioration, respectively.
CONCLUSIONS: The minimal clinically important difference represents the smallest
change in an outcome measure that is meaningful to patients. Our estimates for
the Burke-Fahn-Marsden Dystonia Rating Scale and the 36-Item Short-Form Health
Survey may allow more reliable judgment of the clinical relevance of different
treatments for segmental and generalized isolated dystonia. © 2020 The Authors.
Movement Disorders published by Wiley Periodicals, Inc. on behalf of
International Parkinson and Movement Disorder Society. BACKGROUND: The Dyskinesia Impairment Scale (DIS) is a new assessment scale for
dystonia and choreoathetosis in children and youth with dyskinetic cerebral
palsy. Today, the Burke-Fahn-Marsden Dystonia Rating Scale (BFM) is mostly used
to assess dystonia in children with inherited dystonia. The aim of this study
was to assess reliability and validity of the DIS in children and youth with
inherited or idiopathic dystonia.
METHODS: Reliability was measured by (1) the intraclass correlation coefficients
(ICCs) for inter-rater and test-retest reliability, as well as (2) standard
error of measurement (SEM) and minimal detectable difference (MDD). For
concurrent validity of the DIS-dystonia subscale, the BFM was administered.
RESULTS: In total, 11 males and 9 females (median age 16 years and 7 months,
range 6 to 24 years) were included. For inter-rater reliability, the ICCs for
the DIS total score and the dystonia and choreoathetosis subscale scores were
0.83, 0.87, and 0.71, respectively. For test-retest reliability, the ICCs for
the DIS total score and the dystonia and choreoathetosis subscale scores were
0.95, 0.88, and 0.93, respectively. The SEM and MDD for the total DIS were 3.98%
and 11.04%, respectively. The Spearman correlation coefficient between the
dystonia subscale and the BFM was 0.88 (p < 0.01).
CONCLUSIONS: Good to excellent inter-rater, test-retest reliability, and
validity were found for the total DIS and the dystonia subscale. The
choreoathetosis subscale showed moderate inter-rater reliability and excellent
test-retest reliability. The DIS may be a promising tool to assess dystonia and
choreoathetosis in children and young adults with inherited or idiopathic
dystonia. BACKGROUND: Mutations in TUBB4A are associated with a wide phenotypic spectrum
including generalized dystonia with whispering dysphonia (DYT-TUBB4A).
METHODS: We report the case of a 44-year-old patient with DYT-TUBB4A with a
clinical presentation of disabling progressive dystonia, with a prominent
laryngeal, cervical and facial involvement.
RESULTS: Bipallidal deep brain stimulation (DBS) resulted in a 55% reduction of
dystonia severity assessed by the Burke-Fahn-Marsden scale score 6 months after
surgery. The effect was obvious on the cervical and facial components of
dystonia.
CONCLUSION: We suggest that bipallidal DBS should be considered in patients with
disabling dystonia related to TUBB4A variants. |
Is methotrexate used for the treatment of Rheumatoid Arthritis (RA)? | Yes, methotrexate is effective for the treatment of Rheumatoid Arthritis. MTX can be administered in combination with other chemotherapeutic agents or as mono-therapy. Response rate to MTX is more than 90%. MTX therapy is associated with improved survival of RA patients. | Methotrexate (MTX) is currently under study for use in juvenile rheumatoid
arthritis. One complication of MTX is hepatotoxicity. Although liver function
tests may be abnormal with its use, in this setting they do not correlate well
with the development of hepatic fibrosis. Periodic liver biopsy is required to
monitor for the hepatotoxic changes secondary to MTX. We describe and discuss
the case of a 17-year-old woman who developed evidence of hepatic fibrosis after
3 years of MTX therapy. Twenty-nine patients participated in a prospective study of the safety and
efficacy of oral methotrexate in the treatment of refractory rheumatoid
arthritis. Patients received a mean dosage of 12.4 mg weekly over a mean
duration of 29.1 months. All patients had liver biopsies at baseline, 2 years,
and annually thereafter. Patients improved significantly by all clinical
measures of efficacy after 1 month; maximum improvement tended to occur after
approximately 6 months of therapy. Radiographs showed improvement of erosive
disease in 7 of 11 patients measured. There was a significant reduction in mean
prednisone dosage. Four patients required an increase in the dosage of
methotrexate after prolonged therapy, because of declining clinical response.
Toxicity was noted at some time in 26 of 29 patients (90%), but reactions
universally became mild and tolerable after adjustment of the dosage. No
significant hepatotoxicity was found in 60 sequential liver biopsies, although
elevated transaminase levels were noted at some time in 20 of 29 patients (70%). The use of methotrexate in rheumatoid arthritis is reviewed. Methotrexate, a
folic acid antagonist, is sometimes employed in an attempt to symptomatically
control patients whose disease does not respond adequately to conventional
therapies. Systemic administration of 7.5-15 mg/wk in a "pulse" fashion appears
to be effective without precipitating severe adverse effects. However, concern
over potentially serious side effects and a lack of well-controlled clinical
trials have limited its use to severe, refractory disease. Further studies are
needed before its role in rheumatoid arthritis can justifiably be expanded. Low dose pulse methotrexate (MTX) has become a widely used therapy for
rheumatoid arthritis (RA) because of its good response rate profile. With the
increased use of MTX, reports of opportunistic infections associated with MTX
therapy have appeared. Fourteen cases of pneumocystis carinii (PC) pneumonia in
patients receiving low dose MTX have been previously reported. Yet, no case of
PC pneumonia associated with low dose MTX has so far been reported in Japan. We
report the first case in Japan of PC pneumonia occurring in a patient with
rheumatoid vasculitis who was receiving low dose MTX. A 70-year old woman with
13 year history of RA presented with 3-day history of rapidly aggravating
dyspnea, dry cough and fever. She had been receiving MTX 7.5 mg/week for 2.5
months because of her vasculitis symptoms. She had also been receiving
prednisolone 7.5 mg/day which had been successfully tapered from an initial dose
of 15 mg/day. At the time of her presentation with respiratory symptoms, all of
her vasculitis symptoms had been alleviated. A chest radiograph revealed diffuse
interstitial shadowing bilaterally and bilateral hilar and right lower lung
field infiltrates. Her arterial blood gas showed severe hypoxemia (PaO2 27.7
torr). Polymerase chain reaction assay of bronchoalveolar lavage fluid showed
PC. Although the patient required ventilatory support for 9 days, she was
successfully treated with trimethoprime-sulphamethoxazole and methylprednisolone
pulse therapy. Eight months later, the patient was well with no evidence of
vasculitis or respiratory symptoms. A number of studies show the efficacy of methotrexate (MTX) for rheumatoid
arthritis (RA) in general. However, is there any reason to single this drug out
for early RA? Mechanistically, it probably works differently in RA than in
cancer, at least in part. Thus, in addition to dihydrofolate reductase-related
effects, MTX inhibits aminoimidazocarboxamide transformylase, decreases
leukotriene B4 production, and increases adenosine release at concentrations
achieved with low-dose MTX regimens. Clinically, it is well tolerated over
relatively long periods. Further, a recent meta-analysis of radiology studies
shows that MTX compares favorably with intramuscular gold and is better than
azathioprine. Toxicity remains a concern in treating early RA, particularly as
pulmonary "hypersensitivity reactions" continue (1% to 7.6%), infections (both
fungal and perioperative) are documented, and more cirrhosis is found. With all
of the above in mind, the use of MTX seems reasonable but not necessarily
uniformly appropriate and not yet proved for early RA. Studies of MTX in early
RA, particularly in combination with other drugs, are only beginning. Increasingly, methotrexate (MTX) and sulphasalazine (SASP) are used initially
for second-line therapy of rheumatoid arthritis (RA). Although SASP and MTX are
commonly used, the mechanism(s) by which these drugs control the inflammation
that characterizes RA have remained obscure. Results from my laboratory indicate
that these agents share a mode of action; the anti-inflammatory effects of both
SASP and MTX are due, in both in vitro and in vivo studies, to their capacity to
enhance adenosine release at inflamed sites. This mode of action suggests that
the development of agents that directly alter adenosine metabolism may lead to
new, more effective and safer antirheumatic drugs than those currently
available. OBJECTIVES: The folate antagonist methotrexate (MTX) has become established as
the most commonly used disease-modifying anti-rheumatic drug (DMARD) in the
treatment of rheumatoid arthritis (RA) but is commonly discontinued due to
adverse effects. Adverse effects are thought to be mediated via folate
antagonism. In this paper we summarize the current data on the use of folates as
a supplement to MTX use in RA for the prevention of adverse effects and as a
potential modulator of cardiovascular risk, and propose guidelines for standard
practice.
METHODS: A Medline search was performed using the search terms "methotrexate",
"folic acid", "folinic acid", "folate" and "homocysteine". Literature relevant
to the use of folates as a supplement to MTX in the treatment of RA was reviewed
and other papers referred to as references were explored.
RESULTS: The use of supplemental folates, including folic and folinic acid, in
RA patients treated with MTX has been shown to improve continuation rates by
reducing the incidence of liver function test abnormalities and gastrointestinal
intolerance. Folate supplements do not appear to significantly reduce the
effectiveness of MTX in the treatment of RA. Furthermore, supplemental folic
acid offsets the elevation in plasma homocysteine associated with the use of
MTX. This may in turn reduce the risk of cardiovascular disease, which is
over-represented amongst patients with RA, and for which hyperhomocysteinaemia
is now recognized as an independent risk factor.
CONCLUSIONS: We propose that folic acid supplements be prescribed routinely to
all patients receiving MTX for the treatment of RA. We recommend a pragmatic
dosing schedule of 5 mg of oral folic acid given on the morning following the
day of MTX administration. Methotrexate (MTX) has been the anchor treatment in rheumatoid arthritis (RA)
over the last 15 years, and is used in combination with biologic agents to
enhance efficacy over the last decade or so. The safety profile of MTX has been
studied over 25 years with very few clinically important adverse events in the
weekly low-doses used for RA treatment. The importance of MTX in earlier and
more aggressive management of RA patients cannot be overstated. MTX courses show
some of the longest continuation rates reported in clinical medicine, due to
both effectiveness and safety. The safety profile of MTX indicates that it is
among the safest of any mediation used for the treatment of any arthritis.
Better information on the effectiveness and safety of weekly-low dose MTX should
be communicated to all health professionals involved in the management of RA
patients. Methotrexate (MTX) is currently the most frequently used drugs in the treatment
of rheumatoid arthritis (RA). The drug had been synthesized in 1948 and first
tests to treat patients with psoriasis and RA were published in 1951. However,
until the 1980s there was only limited use of MTX in the treatment of RA. Since
the 1990s MTX is the disease-modifying antirheumatic drug (DMARD) of first
choice for the treatment of RA in most countries worldwide. By definition,
DMARDs in RA are those compounds for which an inhibiting effect on radiographic
progression has been demonstrated. Several combinations of DMARDs have been
tested, most commonly with MTX as the anchor drug. Regarding the route of
administration of MTX there is some evidence that the parenteral route, most
often performed subcutaneously, has some additional benefits over the oral
route. In MTX monotherapy, dosages up to 30 mg/week are now used. There are now
three main combinations that are playing an important role: MTX + sulfasalazine
(SSZ) + hydroxychloroquine, MTX + leflunomide (LEF), and MTX + biologics such as
antitumour necrosis factor (anti-TNF) and other new compounds which block the
interleukin 6 (IL6) receptor or T-cell activation and delete B cells. Regarding
clinical efficacy, MTX monotherapy has performed almost similarly well in
comparison with biologic mono-therapy, both usually combined with
glucocorticoids. However, structural damage is usually inhibited to a
significantly greater degree with the biologics. The combination of MTX with
biologics has proven superior to either agent alone in all aspects. Current
strategic regimens which concentrate on systematic ways to bring patients into
remission all include MTX as first choice. The objective of this review is to update the recommendations of the 2010
Italian Consensus on the use of methotrexate (MTX) in rheumatoid arthritis (RA)
and other rheumatic diseases. The literature published between 2008 and 2012 was
systematically reviewed and updated recommendations on MTX use in rheumatic
diseases, particularly RA, were formulated. These recommendations were approved
by a panel of expert Italian Rheumatologists. A total of 10,238 references were
identified, among which 70 studies were selected for critical evaluation.
Sufficient evidence had accumulated to warrant changes to several of the
recommendations in the new version. A new recommendation for patients with RA
who are in MTX-induced clinical remission was also proposed and approved by the
panel. Updated recommendations for the use of MTX in patients with RA or other
rheumatologic disease are proposed. The association of rheumatoid arthritis (RA) and immune thrombocytopenic purpura
(ITP) has been reported rarely. Methotrexate, which is used for RA treatment,
causes thrombocytopenia. Therefore, in medical practice, physicians avoid using
methotrexate for RA in patients who have both RA and ITP. Here, we report an RA
case that also had ITP, which did not decrease in platelet count after
methotrexate therapy. A 50-year-old woman was diagnosed with diabetes mellitus
in 1990, RA in 1995, and ITP in 2000. She had received hydroxychloroquine for
more than 5 years. She was treated with prednisolone 16 mg/daily between 2006
and 2007, but she discontinued this therapy because of weight gain. Laboratory
findings were not remarkable, except for thrombocytopenia. We started
methotrexate therapy 10 mg per week for treatment of RA, and hydroxychloroquine
therapy was stopped due to nonresponse. The methotrexate dose was increased up
to 15 mg/week. Her complete blood cell count was monitored frequently. We did
not observe any decrease in platelet count, while active arthritis symptoms of
the patient were relieved. This case shows that methotrexate may be used in
patients diagnosed with RA that is associated with ITP under strict monitoring. BACKGROUND: Treatment with methotrexate (MTX) in patients with rheumatoid
arthritis (RA) leads to decreased total immunoglobulin (Ig) levels and impairs
vaccine-specific IgG antibody levels following pneumococcal vaccination. The
mechanisms by which MTX exerts these effects in RA are unknown. We aimed to
evaluate whether MTX reduces vaccine-specific serum Ig levels and their
functionality in RA patients following vaccination with pneumococcal conjugate
vaccine, and if numbers of antigen-specific circulating plasmablasts are
affected.
METHODS: Ten patients with RA on MTX and 10 RA patients without disease
modifying anti-rheumatic drug (DMARD) were immunized with a dose 13-valent
pneumococcal conjugate vaccine (Prevenar13). Circulating plasmablasts producing
total IgG and IgA as well as specific IgG and IgA against two pneumococcal
capsular serotypes (6B and 23F) were enumerated using ELISPOT 6days after
vaccination. IgG levels against both these serotypes were determined with ELISA
before and 4-6weeks after vaccination. Positive antibody response was defined as
⩾2-fold increase of pre-vaccination antibody levels. The functionality of
vaccine specific antibodies to serotype 23F was evaluated by measuring their
ability to opsonize bacteria using opsonophagocytic assay (OPA) in 4 randomly
chosen RA patients on MTX and 4 RA patients without DMARD.
RESULTS: After vaccination, RA patients on MTX showed significant increase in
pre- to postvaccination antibody levels for 6B (p<0.05), while patients without
DMARD had significant increases for both 6B and 23F (p<0.05 and p<0.01,
respectively). Only 10% of RA on MTX and 40% of RA patients without DMARD showed
positive post-vaccination antibody responses for both serotypes. Increased
opsonizing ability after vaccination was detected in 1 of 4 RA patients on MTX
and 3 of 4 patients on RA without DMARD. However, numbers of circulating total
and vaccine-specific IgG- or IgA-producing plasmablasts did not differ between
RA patients with or without MTX.
CONCLUSIONS: MTX treatment in RA leads to reduced vaccine-specific antibody
responses and their functionality compared to untreated RA following
pneumococcal vaccination using polysaccharide-protein conjugate vaccine.
However, since there was no reduction in numbers of circulating total or
vaccine-specific antibody-producing plasmablasts after vaccination this effect
is probably not due to reduced activation of B cells in lymphoid tissue.
CLINICAL TRIAL REGISTRATION: NCT02240888. In rheumatoid arthritis (RA) treatment, the concomitant use of methotrexate has
been shown to reduce the incidence of antibodies to infliximab (ATI), on the
other hand, it is unclear whether azathioprine can reduce ATI production. We
enrolled a total of 10 Japanese adult patients with RA who were treated with
infliximab concomitantly with methotrexate or azathioprine. Serum concentrations
of infliximab and ATI of these patients were measured. The mean serum infliximab
concentrations was 1.6±1.3 μg/ml in patients with methotrexate and 1.0±0.5 μg/ml
in patients with azathioprine. Serum ATI concentrations were below the limit of
quantitation in 4 of 5 patients in each group. The results from the present
study suggest that azathioprine suppresses ATI production. Methotrexate has been used in treatment of rheumatoid arthritis (RA) since the
1980s and to this day is often the first line medication for RA treatment. In
this review, we examine multiple hypotheses to explain the mechanism of
methotrexate efficacy in RA. These include folate antagonism, adenosine
signaling, generation of reactive oxygen species (ROS), decrease in adhesion
molecules, alteration of cytokine profiles, and polyamine inhibition amongst
some others. Currently, adenosine signaling is probably the most widely accepted
explanation for the methotrexate mechanism in RA given that methotrexate
increases adenosine levels and on engagement of adenosine with its extracellular
receptors an intracellular cascade is activated promoting an overall
anti-inflammatory state. In addition to these hypotheses, we examine the
mechanism of methotrexate in RA from the perspective of its adverse effects and
consider some of the newer genetic markers of methotrexate efficacy and toxicity
in RA. Lastly, we briefly discuss the mechanism of additive methotrexate in the
setting of TNF-α inhibitor treatment of RA. Ultimately, finding a clear
explanation for the pathway and mechanism leading to methotrexate efficacy in
RA, there may be a way to formulate more potent therapies with fewer side
effects. Conflict of interest statement: CONFLICT OF INTEREST: SS has received speaking
fees from Chugai Pharmaceutical, Eisai, Bristol–Myers K.K, Asahikasei Pharma
Corp, Pfizer Japan, and consultant fees from Asahikasei Pharma Corp. TT has
received research grants from Astellas Pharma Inc, Bristol–Myers K.K., Chugai
Pharmaceutical Co, Ltd., Daiichi Sankyo Co., Ltd., Takeda Pharmaceutical Co.,
Ltd., Teijin Pharma Ltd., AbbVie GK, Asahikasei Pharma Corp., Mitsubishi Tanabe
Pharma Co., Pfizer Japan Inc., and Taisho Toyama Pharmaceutical Co., Ltd., Eisai
Co., Ltd., AYUMI Pharmaceutical Corporation, speaking fees from AbbVie GK.,
Bristol–Myers K.K., Chugai Pharmaceutical Co,. Ltd., Mitsubishi Tanabe Pharma
Co., Pfizer Japan Inc., and Astellas Pharma Inc, and Daichi Sankyo Co., Ltd, and
consultant fees from Astra Zeneca K.K., Eli Lilly Japan K.K., Novartis Pharma
K.K., Mitsubishi Tanabe Pharma Co., Abbvie GK, Nipponkayaku Co., Ltd, Janssen
Pharmaceutical K.K., Astellas Pharma Inc. Despite the introduction of numerous biologic agents for the treatment of
rheumatoid arthritis (RA) and other forms of inflammatory arthritis, low-dose
methotrexate therapy remains the gold standard in RA therapy. Methotrexate is
generally the first-line drug for the treatment of RA, psoriatic arthritis and
other forms of inflammatory arthritis, and it enhances the effect of most
biologic agents in RA. Understanding the mechanism of action of methotrexate
could be instructive in the appropriate use of the drug and in the design of new
regimens for the treatment of RA. Although methotrexate is one of the first
examples of intelligent drug design, multiple mechanisms potentially contribute
to the anti-inflammatory actions of methotrexate, including the inhibition of
purine and pyrimidine synthesis, transmethylation reactions, translocation of
nuclear factor-κB (NF-κB) to the nucleus, signalling via the Janus kinase
(JAK)-signal transducer and activator of transcription (STAT) pathway and nitric
oxide production, as well as the promotion of adenosine release and expression
of certain long non-coding RNAs. |
What methodology does the FoundationOne CDx test use? | FoundationOne CDx is a next generation sequencing (NGS) based test. | BACKGROUND: Immunotherapy has demonstrated encouraging clinical benefits in
patients with advanced breast carcinomas and Programmed death ligand 1 (PD-L1)
expression has been proposed as an immunotherapy biomarker. Challenges with
current PD-L1 testing exist and tumor mutation burden (TMB) is emerging as a
biomarker to predict clinical response to immunotherapy in melanoma and
non-small cell lung cancer patients. However, TMB has not been well
characterized in breast carcinomas.
METHODS: The study cohort included 62 advanced breast cancer patients (13
primary and 49 metastatic). Genetic alterations and TMB were determined by
FoundationOne CDx next generation sequencing (NGS) and the association with
clinicopathologic features was analyzed.
RESULTS: High TMB was observed in a relatively low frequency (3/62, 4.8%). TMB
levels were positively associated tumor infiltrating lymphocytes and
significantly higher TMB was observed in breast carcinomas with DNA damage
repair gene mutation(s). There was no significant association between TMB levels
and other analyzed clinicopathologic characteristics.
CONCLUSIONS: Our data indicate the importance of DNA damage repair proteins in
maintaining DNA integrity and immune reaction and breast carcinoma patients with
DDR mutation may benefit from immunotherapy. |
Which R/bioconductor package exists for discovery of intergenic transcripts? | To increase the power of transcript discovery from large collection of RNA-seq data sets, a novel '1-Step' approach named pooling RNA-Seq and Assembling Models (PRAM) has been developed that build transcript models from pooled RNA- sequencing data sets. PRAM is implemented as an R/Bioconductor package. | Publicly available RNA-seq data is routinely used for retrospective analysis to
elucidate new biology. Novel transcript discovery enabled by joint analysis of
large collections of RNA-seq data sets has emerged as one such analysis. Current
methods for transcript discovery rely on a '2-Step' approach where the first
step encompasses building transcripts from individual data sets, followed by the
second step that merges predicted transcripts across data sets. To increase the
power of transcript discovery from large collections of RNA-seq data sets, we
developed a novel '1-Step' approach named Pooling RNA-seq and Assembling Models
(PRAM) that builds transcript models from pooled RNA-seq data sets. We
demonstrate in a computational benchmark that 1-Step outperforms 2-Step
approaches in predicting overall transcript structures and individual splice
junctions, while performing competitively in detecting exonic nucleotides.
Applying PRAM to 30 human ENCODE RNA-seq data sets identified unotated
transcripts with epigenetic and RAMPAGE signatures similar to those of recently
annotated transcripts. In a case study, we discovered and experimentally
validated new transcripts through the application of PRAM to mouse hematopoietic
RNA-seq data sets. We uncovered new transcripts that share a differential
expression pattern with a neighboring gene Pik3cg implicated in human
hematopoietic phenotypes, and we provided evidence for the conservation of this
relationship in human. PRAM is implemented as an R/Bioconductor package. |
What is the mechanism of action of idasanutlin? | Idasanutlin is a small-molecule inhibitor of MDM2, a negative regulator of tumor suppressor p53. | BACKGROUND: Venetoclax, a small molecule BH3 mimetic which inhibits the
anti-apoptotic protein Bcl-2, and idasanutlin, a selective MDM2 antagonist, have
both shown activity as single-agent treatments in pre-clinical and clinical
studies in acute myeloid leukemia (AML). In this study, we deliver the rationale
and molecular basis for the combination of idasanutlin and venetoclax for
treatment of p53 wild-type AML.
METHODS: The effect of idasanutlin and venetoclax combination on cell viability,
apoptosis, and cell cycle progression was investigated in vitro using
established AML cell lines. In vivo efficacy was demonstrated in subcutaneous
and orthotopic xenograft models generated in female nude or non-obese
diabetic/severe combined immunodeficiency (NOD/SCID) mice. Mode-of-action
analyses were performed by means of cell cycle kinetic studies, RNA sequencing
as well as western blotting experiments.
RESULTS: Combination treatment with venetoclax and idasanutlin results in
synergistic anti-tumor activity compared with the respective single-agent
treatments in vitro, in p53 wild-type AML cell lines, and leads to strongly
superior efficacy in vivo, in subcutaneous and orthotopic AML models. The
inhibitory effects of idasanutlin were cell-cycle dependent, with cells
arresting in G1 in consecutive cycles and the induction of apoptosis only
evident after cells had gone through at least two cell cycles. Combination
treatment with venetoclax removed this dependency, resulting in an acceleration
of cell death kinetics. As expected, gene expression studies using RNA
sequencing showed significant alterations to pathways associated with p53
signaling and cell cycle arrest (CCND1 pathway) in response to idasanutlin
treatment. Only few gene expression changes were observed for venetoclax
treatment and combination treatment, indicating that their effects are mediated
mainly at the post-transcriptional level. Protein expression studies
demonstrated that inhibition of the anti-apoptotic protein Mcl-1 contributed to
the activity of venetoclax and idasanutlin, with earlier inhibition of Mcl-1 in
response to combination treatment contributing to the superior combined
activity. The role of Mcl-1 was confirmed by small hairpin RNA gene knockdown
studies.
CONCLUSIONS: Our findings provide functional and molecular insight on the
superior anti-tumor activity of combined idasanutlin and venetoclax treatment in
AML and support its further exploration in clinical studies. PURPOSE: Idasanutlin, a selective small-molecule MDM2 antagonist in phase 3
testing for refractory/relapsed AML, is a non-genotoxic oral p53 activator. To
optimize its dosing conditions, a number of clinical pharmacology
characteristics were examined in this multi-center trial in patients with
advanced solid tumors.
METHOD: This was an open-label, single-dose, crossover clinical pharmacology
study investigating the effects of strong CYP3A4 inhibition with posaconazole
(Part 1), two new oral formulations (Part 2), as well as high-energy/high-fat
and low-energy/low-fat meals (Part 3) on the relative bioavailability of
idasanutlin. After completing Part 1, 2, or 3, patients could have participated
in an optional treatment with idasanutlin. Clinical endpoints were
pharmacokinetics (PK), pharmacodynamics (PD) of MIC-1 elevation (Part 1 only),
and safety/tolerability.
RESULTS: The administration of posaconazole 400 mg BID × 7 days with idasanutlin
800 mg resulted in a slight decrease (7%) in Cmax and a modest increase (31%) in
AUC for idasanutlin, a marked reduction in Cmax (~ 60%) and AUC0 (~ 50%) for M4
metabolite, and a minimal increase (~ 24%) in serum MIC-1 levels. Cmax and AUC
were both 45% higher for the SDP formulation. While the low-fat meal caused a
less than 20% increase in all PK exposure parameters with the 90% CI values just
outside the upper end of the equivalence criteria (80-125%), the high-fat meal
reached bioequivalence with dosing under fasting.
CONCLUSION: In patients with solid tumors, multiple doses of posaconazole, a
strong CYP3A4 inhibitor, minimally affected idasanutlin PK and PD without
clinical significance. The SDP formulation improved rBA/exposures by ~ 50%
without major food effect. PURPOSE: Idasanutlin, a selective small-molecule MDM2 antagonist in phase 3
testing for refractory/relapsed AML, is a non-genotoxic oral p53 activator. The
aim of this analysis is to examine the potential of idasanutlin to prolong the
corrected QT (QTc) interval by evaluating the relationship between plasma
idasanutlin concentration and QTc interval.
METHOD: Intensive plasma concentration QTc interval data were collected at the
same timepoints, from three idasanutlin (RO5503781) phase 1 studies in patients
with solid tumors and AML. QTc data in absolute values and changes from baseline
(Δ) were analyzed for a potential association with plasma idasanutlin
concentrations with a linear mixed effect model. Categorical analysis was also
performed.
RESULTS: A total of 282 patients were exposed to idasanutlin and had at least
one observation of QTc and idasanutlin plasma concentration. There was no
apparent increase of QTcF or ΔQTcF in a wide idasanutlin plasma concentration
range, even at concentrations exceeding the exposure matching the dose adopted
in the ongoing phase 3 study (300-mg BID). Categorical analysis did not detect a
potential signal of QT prolongation.
CONCLUSION: The concentration-QTc analysis indicates that idasanutlin does not
prolong the QT interval within the targeted concentration range currently in
consideration for clinical development. Venetoclax (ABT-199) and idasanutlin (RG7388) are efficient anticancer drugs
targeting two essential apoptosis markers, Bcl-2 and MDM2, respectively. Recent
studies have shown that the combination of these two drugs leads to remarkable
enhancement of anticancer efficacy, both in vitro and in vivo. In an attempt to
disclose the relationships of their protein targets, competitive affinity-based
proteome profiling coupled with bioimaging was employed to characterize their
protein targets in the same cancer cell line and tumor tissue. A series of
protein hits, including ITPR1, GSR, RER1, PDIA3, Apoa1, and Tnfrsf17 were
simultaneously identified by pull-down/LC-MS/MS with the two sets of
affinity-based probes. Dual imaging was successfully carried out, with the
simultaneous detection of Bcl-2 and MDM2 expression in various cancer cells.
This could facilitate the novel diagnostic and therapeutic strategies of dual
targeting of Bcl-2/MDM2. A concise asymmetric synthesis has been developed to prepare idasanutlin, a
small molecule MDM2 antagonist. Idasanutlin is currently being investigated as a
potential treatment for various solid tumors and hematologic maligcies. The
highly congested pyrrolidine core, containing four contiguous stereocenters, was
constructed via a Cu(I)/(R)-BINAP catalyzed [3+2]-cycloaddition reaction. This
optimized copper(<small>I</small>)-catalyzed process has been used to produce
more than 1500 kg of idasanutlin. The manufacturing process will be described,
highlighting the exceptionally selective and consistent
cycloaddition/isomerization/hydrolysis sequence. The excellent yields, short
cycle times and reduction in waste streams result in a sustainable production
process with low environmental impact. The protein p53 protects the organism against carcinogenic events by the
induction of cell cycle arrest and DNA repair program upon DNA damage. Virtually
all cancers inactivate p53 either by mutations/deletions of the TP53 gene or by
boosting negative regulation of p53 activity. The overexpression of MDM2 protein
is one of the most common mechanisms utilized by p53wt cancers to keep p53
inactive. Inhibition of MDM2 action by its antagonists has proved its anticancer
potential in vitro and is now tested in clinical trials. However, the prolonged
treatment of p53wt cells with MDM2 antagonists leads to the development of
secondary resistance, as shown first for Nutlin-3a, and later for three other
small molecules. In the present study, we show that secondary resistance occurs
also after treatment of p53wt cells with idasanutlin (RG7388, RO5503781), which
is the only MDM2 antagonist that has passed phase II and entered phase III
clinical trials, so far. Idasanutlin strongly activates p53, as evidenced by the
induction of p21 expression and potent cell cycle arrest in all the three cell
lines tested, i.e., MCF-7, U-2 OS, and SJSA-1. Notably, apoptosis was induced
only in SJSA-1 cells, while MCF-7 and U-2 OS cells were able to restore the
proliferation upon the removal of idasanutlin. Moreover, idasanutlin-treated U-2
OS cells could be cultured for long time periods in the presence of the drug.
This prolonged treatment led to the generation of p53-mutated resistant cell
populations. This resistance was generated de novo, as evidenced by the
utilization of monoclonal U-2 OS subpopulations. Thus, although idasanutlin
presents much improved activities compared to its precursor, it displays the
similar weaknesses, which are limited elimination of cancer cells and the
generation of p53-mutated drug-resistant subpopulations. BACKGROUND AND OBJECTIVE: Alterations in gene expressions are often due to
epigenetic modifications that can have a significant influence on cancer
development, growth, and progression. Lately, histone deacetylase inhibitors
(HDACi) such as suberoylanilide hydroxamic acid (SAHA, or vorinostat, MK0683)
have been emerging as a new class of drugs with promising therapeutic benefits
in controlling cancer growth and metastasis. The small molecule RG7388
(idasanutlin, R05503781) is a newly developed inhibitor that is specific for an
oncogene-derived protein called MDM2, which is also in clinical trials for the
treatment of various types of cancers. These two drugs have shown the ability to
induce p21 expression through distinct mechanisms in MCF-7 and LNCaP cells,
which are reported to have wild-type TP53. Our understanding of the molecular
mechanism whereby SAHA and RG7388 can induce cell cycle arrest and trigger cell
death is still evolving. In this study, we performed experiments to measure the
cell cycle arrest effects of SAHA and RG7388 using MCF-7 and LNCaP cells.
MATERIALS AND METHODS: The cytotoxicity, cell cycle arrest, and
apoptosis/necroptosis effects of the SAHA and RG7388 treatments were assessed
using the Trypan Blue dye exclusion (TBDE) method,
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay,
fluorescence assay with DEVD-amc substrate, and immunoblotting methods.
RESULTS: The RG7388 treatment was able to induce cell death by elevating
p21WAF1/CIP1 through inhibition of MDM2 in LNCaP, but not in MCF-7 cells, even
though there was evidence of p53 elevation. Hence, we suspect that there is some
level of uncoupling of p53-mediated transcriptional induction of p21WAF1/CIP1 in
MCF-7 cells.
CONCLUSION: Our results from MCF-7 and LNCaP cells confirmed that SAHA and
RG7388 treatments were able to induce cell death via a combination of cell cycle
arrest and cytotoxic mechanisms. We speculate that our findings could lead to
the development of newer treatments for breast and prostate cancers with drug
combinations including HDACi. Maligt rhabdoid tumors (MRT) are highly aggressive pediatric cancers that
respond poorly to current therapies. In this study, we screened several MRT cell
lines with large-scale RNAi, CRISPR-Cas9, and small-molecule libraries to
identify potential drug targets specific for these cancers. We discovered MDM2
and MDM4, the canonical negative regulators of p53, as significant
vulnerabilities. Using two compounds currently in clinical development,
idasanutlin (MDM2-specific) and ATSP-7041 (MDM2/4-dual), we show that MRT cells
were more sensitive than other p53 wild-type cancer cell lines to inhibition of
MDM2 alone as well as dual inhibition of MDM2/4. These compounds caused
significant upregulation of the p53 pathway in MRT cells, and sensitivity was
ablated by CRISPR-Cas9-mediated inactivation of TP53. We show that loss of
SMARCB1, a subunit of the SWI/SNF (BAF) complex mutated in nearly all MRTs,
sensitized cells to MDM2 and MDM2/4 inhibition by enhancing p53-mediated
apoptosis. Both MDM2 and MDM2/4 inhibition slowed MRT xenograft growth in vivo,
with a 5-day idasanutlin pulse causing marked regression of all xenografts,
including durable complete responses in 50% of mice. Together, these studies
identify a genetic connection between mutations in the SWI/SNF
chromatin-remodeling complex and the tumor suppressor gene TP53 and provide
preclinical evidence to support the targeting of MDM2 and MDM4 in this
often-fatal pediatric cancer. SIGNIFICANCE: This study identifies two targets,
MDM2 and MDM4, as vulnerabilities in a deadly pediatric cancer and provides
preclinical evidence that compounds inhibiting these proteins have therapeutic
potential. Purpose MDM2 is a negative regulator of the tumor suppressor p53. RO6839921 is
an inactive pegylated prodrug of idasanutlin, an MDM2 antagonist, developed for
intravenous administration. On cleavage by plasma esterases, the active
principle (AP = idasanutlin) is released. This phase 1 study investigated the
safety, pharmacokinetics, and pharmacodynamics of RO6839921 in patients with
advanced solid tumors (NCT02098967). Methods Patients were evaluated on a 5-day
dosing schedule every 28 days. Dose escalation used the Bayesian new continual
reassessment model. Accelerated dose titration was permitted until grade ≥2
drug-related AEs were observed. The target DLT rate to define the MTD was
16-25%. p53 activation was assessed by measuring macrophage inhibitory
cytokine-1 (MIC-1). Results Forty-one patients received 14-120 mg AP; 39 were
DLT evaluable. The MTD was 110-mg AP (8% DLT rate), whereas 120-mg AP had a 44%
DLT rate. DLTs were neutropenia, thrombocytopenia, and stridor. The most common
treatment-related AEs (≥30%) were nausea, fatigue, vomiting, and
thrombocytopenia. Pharmacokinetic analyses indicated rapid conversion of prodrug
to AP and an approximately linear and dose-proportional dose-exposure
relationship, with a 2-fold increase in exposure between Days 1 and 5 of AP.
MIC-1 increases were exposure dependent. Stable disease was observed in 14
patients (34%). Conclusions RO6839921 showed reduced pharmacokinetic exposure
variability and a safety profile comparable with that of oral idasanutlin.
Although this study indicated that RO6839921 could be administered to patients,
the results did not provide sufficient differentiation or improvement in the
biologic or safety profile compared with oral idasanutlin to support continued
development. In acute myeloid leukemia (AML), TP53 mutations and dysregulation of wild-type
p53 is common and supports an MDM2 antagonist as a therapy. RO6839921 is an
inactive pegylated prodrug of the oral MDM2 antagonist idasanutlin (active
principle [AP]) that allows for IV administration. This phase 1 monotherapy
study evaluated the safety, pharmacokinetics, and pharmacodynamics of RO6839921
in patients with AML. Primary objectives identified dose-limiting toxicities
(DLTs) and maximum tolerated dose (MTD). Secondary objectives assessed
pharmacokinetic, pharmacodynamic, and antileukemic activity. A total of 26
patients received 120-300 mg AP of idasanutlin. The MTD was 200 mg, with DLTs at
250 (2/8 patients) and 300 mg (2/5). Treatment-related adverse events in >20% of
patients were diarrhea, nausea, vomiting, decreased appetite, and fatigue. Six
deaths (23.1%) occurred, all unrelated to treatment. Pharmacokinetics showed
rapid and near-complete conversion of the prodrug to AP and dose-proportional
exposure across doses. Variability ranged from 30%-47% (22%-54% for
idasanutlin). TP53 was 21 (87.5%) wild-type and 3 mutant (12.5%). The composite
response rate (complete remission [CR], CR with incomplete hematologic
recovery/morphological leukemia-free state [CRi/MLFS], or CR without platelet
recovery [CRp]) was 7.7%. Antileukemic activity (CR, CRi/MLFS, partial response,
hematologic improvement/stable disease) was observed in 11 patients (disease
control rate, 42%): 10/11 were TP53 wild-type; 1 had no sample. p53 activation
was demonstrated by MIC-1 induction and was associated with AP exposure. There
was not sufficient differentiation or improvement in the biologic or safety
profile compared with oral idasanutlin to support continued development of
RO6839921. NCT02098967. Author information:
(1)Departamento de Hematologia, Hospital Universitari i Politècnic La Fe,
València, Spain.
(2)CIBERONC, Instituto Carlos III, Madrid, Spain.
(3)F. Hoffmann-La Roche Ltd, Basel, Switzerland.
(4)Hospital Clinic de Barcelona, IDIBAPS, Barcelona, Spain.
(5)Department of Leukemia, Division of Cancer Medicine, The University of Texas
MD Anderson Cancer Center, Houston, TX, USA.
(6)Department of Hematology and Sciences Oncology, Istituto Scientifico
Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy.
(7)Department of Experimental, Diagnostic and Specialty Medicine Institute of
Hematology & Medical Oncology L & A Seràgnoli, Bologna, Italy.
(8)Hoffmann-La Roche Ltd, Mississauga, ON, Canada.
(9)Serviced'Hématologie, Institut Universitaire du Cancer Toulouse -
Oncopole,Toulouse, France.
(10)Department of Internal Medicine, Universitätsklinikum Carl Gustav Carus,
Dresden, Germany.
(11)Hematology Department, Aix-Marseille University, Institut Paoli-Calmettes,
Marseille, France.
(12)Department of Haematology, The Alfred Hospital & Monash University,
Melbourne, VIC, Australia.
(13)Division of Hematology/Medical Oncology, Department of Internal Medicine,
Seoul National University Hospital, Seoul, South Korea.
(14)Service d'Hématologie Séniors Hôpital Saint-Louis, Assistance Publique -
Hôpitaux de Paris, Université de Paris, Paris, France. BACKGROUND: Despite intensive treatment protocols and recent advances,
neuroblastomas still account for approximately 15% of all childhood cancer
deaths. In contrast with adult cancers, p53 pathway inactivation in
neuroblastomas is rarely caused by p53 mutation but rather by altered MDM2 or
p14ARF expression. Moreover, neuroblastomas are characterised by high
proliferation rates, frequently triggered by pRb pathway dysfunction due to
aberrant expression of cyclin D1, CDK4 or p16INK4a. Simultaneous disturbance of
these pathways can occur via co-amplification of MDM2 and CDK4 or homozygous
deletion of CDKN2A, which encodes both p14ARF and p16INK4a.
METHODS AND RESULTS: We examined whether both single and combined inhibition of
MDM2 and CDK4/6 is effective in reducing neuroblastoma cell viability. In our
panel of ten cell lines with a spectrum of aberrations in the p53 and pRb
pathway, idasanutlin and abemaciclib were the most potent MDM2 and CDK4/6
inhibitors, respectively. No correlation was observed between the genetic
background and response to the single inhibitors. We confirmed this lack of
correlation in isogenic systems overexpressing MDM2 and/or CDK4. In addition,
combined inhibition did not result in synergistic effects. Instead, abemaciclib
diminished the pro-apoptotic effect of idasanutlin, leading to slightly
antagonistic effects. In vivo treatment with idasanutlin and abemaciclib led to
reduced tumour growth compared with single drug treatment, but no synergistic
response was observed.
CONCLUSION: We conclude that p53 and pRb pathway aberrations cannot be used as
predictive biomarkers for neuroblastoma sensitivity to MDM2 and/or CDK4/6
inhibitors. Moreover, we advise to be cautious with combining these inhibitors
in neuroblastomas. Author information:
(1)Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre,
Toronto, ON, Canada. Electronic address: [email protected].
(2)Institute of Hematology "L. and A. Seràgnoli", University Hospital S.
Orsola-Malpighi, Bologna, Italy.
(3)Department of Hematology, Aix-Marseille University, Institut Paoli-Calmettes,
Marseille, France.
(4)Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne
Hospital, and Sir Peter MacCallum Department of Oncology, University of
Melbourne, Melbourne, VIC, Australia.
(5)Division of Hematology, Keck School of Medicine of the University of Southern
California, Los Angeles, CA, United States.
(6)Division of Hematologic Oncology, Segal Cancer Centre, Jewish General
Hospital, Montreal, QC, Canada.
(7)Department of Medical Oncology, Thomas Jefferson University, Philadelphia,
PA, United States.
(8)Department of Medicine, Division of Oncology, New York Medical College,
Valhalla, NY, United States.
(9)Department of Haemato-Oncology, Beatson West of Scotland Cancer Centre,
Glasgow, UK.
(10)Division of Hematology/Medical Oncology, Department of Internal Medicine,
Seoul National University Hospital, Seoul, Republic of Korea.
(11)Department of Hematology, Asan Medical Center, Seoul, Republic of Korea.
(12)Translational Medicine-Oncology, Roche Innovation Center, New York, NY,
United States.
(13)Clinical Pharmacology, Roche Innovation Center, New York, NY, United States.
(14)Clinical Pharmacology, F. Hoffmann-La Roche, Basel, Switzerland.
(15)Product Development Oncology, Genentech, Inc, South San Francisco, CA,
United States.
(16)Department of Biostatistics Oncology, F. Hoffmann-La Roche, Basel,
Switzerland.
(17)Clinical Development Oncology, F. Hoffmann-La Roche, Basel, Switzerland.
(18)Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST),
IRCCS, Meldola, Italy. |
Where is corticosterone synthesized? | Following a stressful event, the hypothalamus-pituitary-adrenal axis mediates the release of the stress hormone cortisol (corticosterone in rodents; CORT). | Arecoline has biomedical importance, but it has untoward side effects on
endocrine functions. The aim is to investigate its role on adrenal activity
under thermal stress by ultrastructural and hormonal parameters in mice. Cold
(4 °C) or heat (37 °C) stress, or arecoline (10 mg/kg body wt), each for 7 days
in cold or heat stress stimulated adrenocortical activity ultrastructurally with
an elevation of corticosterone level. Adrenomedullary activity was suppressed in
cold stress with depletion of catecholamine levels. In heat stress,
adrenomedullary activity was stimulated ultrastructurally with an elevation of
catecholamine levels. Arecoline treatment alone, or in cold or heat stress
suppressed adrenomedullary activity, judged by ultrastructural and hormonal
parameters. Arecoline treatment caused hypoglycemia with an elevation of
glycogen level, but cold or heat stress, or arecoline treatment in thermal
stress caused hyperglycemia, with a fall in glycogen profile. Thus, arecoline in
thermal stress plays a dual role on adrenal function and glucose-glycogen
homeostasis in mice. In full-term elective caesarian sections, fetal flow of adrenal substrate
steroids to products differs by sex, with males (M) in molar equilibrium whereas
females (F) add net molarity and synthesize more cortisol. Using the same
sampling design, paired, full-term, arterial, and venous umbilical cord samples
and intrapartum chart records were obtained at the time of vaginal delivery (N =
167, 85 male) or emergency C-section (N = 38, 22 male). Eight steroids were
quantified by liquid chromatography coupled to tandem mass spectrometry (adrenal
glucocorticoids [cortisol, corticosterone], sequential cortisol precursor
steroids [17-hydroxyprogesterone, 11-deoxycortisol], cortisol and corticosterone
metabolites [cortisone and 11-dehydrocorticosterone], and gonadal steroids
[androstenedione, testosterone]). Fetal sex was not significant in any analytic
models. Going through both phase 1 and phase 2 labor increased fetal adrenal
steroidogenesis and decreased male testosterone relative to emergency C-sections
that do not reach stage 2 of labor (ie, head compressions) and elective
C-sections with no labor. Sum adrenal steroid molarity arriving in venous serum
was almost double the equivalent metric for deliveries without labor. No effects
of operative vaginal delivery were noted. Maternal regional anesthetic
suppressed venous concentrations, and fetal synthesis replaced that steroid.
Approximate molar equivalence between substrate pool depletion and net
glucocorticoid synthesis was seen. Paired venous and arterial umbilical cord
serum has the potential to identify sex differences that underlie antenatal
programming of hypothalamic-pituitary-adrenal axis function in later life.
However, stage 2 labor before the collection of serum, and regional anesthetic
for the mother, mask those sex differences. |
Where is the klotho protein primarily expressed in the body | The klotho protein is primarily expressed in the lungs, kidney, lens, cerebellum, trpc6, renal cells and the brain. | The Klotho mouse is a recently developed model that exhibits phenotypes
resembling human aging. We used this model to investigate sensorineural hearing
loss from the point of view that it may be considered an issue of aging. Using
reverse transcription-polymerase chain reaction, Western blotting and
immunohistochemical staining, we were able to confirm klotho gene transcription
and protein synthesis in the kidney and inner ear. Klotho protein was mainly
expressed in the stria vascularis and spiral ligament of the inner ear and in
the distal convoluted tubule of the kidney, likely serving a common function in
the two organs, i.e., modulating ion transport. The threshold for the auditory
brainstem response was significantly higher in Klotho mice than in wild-type
mice, and wave I latencies were prolonged. On the other hand, Klotho mice
exhibited a normal distribution of I-IV interpeak intervals. No obvious
morphological abnormalities were detected in Klotho mice, although no expression
of Klotho protein was detected, and there was an apparent hearing disorder.
Taken together, these findings suggest that by contributing to the maintece
ion homeostasis in the endolymph, Klotho protein serves as a key mediator of
auditory function. To escape aging and aging-related disorders has been one of mankind's biggest
dreams from the beginning of history. However, our knowledge regarding the
molecular mechanisms of aging has been limited. We recently developed a unique
short lifespan mouse strain in which a single gene mutation caused multiple
aging-related disorders and identified the responsible gene as klotho. The most
characteristic phenotypes seem to be caused by abnormalities in calcium
metabolism. Furthermore, the klotho gene is expressed principally in the
important tissues for calcium homeostasis such as distal tubule cells of the
kidney, choroid plexus in the brain, and the main cells of the parathyroid
gland. Klotho plays a critical role for the regulation of calcium and phosphorus
homeostasis by negatively regulating the synthesis of active Vitamin D. The
deficiency of the klotho gene results in degradation of cells by the activation
of calcium-dependent proteolysis in kidney, lung and heart. Importantly, the
increased activation of calcium-dependent proteolysis occurs in the tissues of
old mice together with the down regulation of klotho gene expression. What is
Klotho protein required for and how does it act? In this review, I will discuss
our working hypotheses on the biological roles and molecular functions of Klotho
protein. Klotho mutant mouse (kl-/-), a mouse model for human aging, exhibits various
phenotypes in a wide range of organs including arteriosclerosis, neural
degeneration, skin and gonadal atrophy, pulmonary emphysema, calcification of
soft tissues, and cognition impairment. Klotho mRNA, however, is expressed only
in brain, kidney, reproductive organs, pituitary gland, and parathyroid gland.
Therefore it remains to be elucidated how lack of Klotho protein in these
limited organs leads to the variety of phenotypes. To shed light on mechanisms
by which Klotho protein acts on distant targets, we examined localization of
Klotho protein in brain, kidney, and reproductive organs, and analyzed brain and
kidney in kl-/- mice searching for changes in target regions in these organs. In
brain, Klotho proteins were localized at choroid plexus, where the proteins were
domitly localized at the apical plasma membrane of ependymal cells. In kl-/-
brain, reduction of synapses was evident in hippocampus, suggesting a role of
Klotho as a humoral factor in cerebrospinal fluid. Klotho proteins in kidney
localized at distal renal tubules. Interestingly, in kl-/-mice, type IIa
Na/phosphate (Pi) cotransporters, which function at the proximal renal tubules
in reabsorption of phosphate ions, were translocated. This suggests that Klotho
protein in kidney is implicated in calcium homeostasis which regulates
localization of type IIa Na/Pi cotransporters via parathyroid hormone (PTH).
Klotho proteins in reproductive organs were expressed only in mature germ cells,
although in kl-/- mice germ cell maturation was arrested at earlier stages.
Thus, Klotho proteins not only function as a humoral factor, but also are
implicated in hormonal regulation, which may explain why mutation of klotho gene
results in a variety of phenotypes. Klotho, a membrane protein mainly expressed in parathyroid glands, kidney, and
choroid plexus, counteracts aging and increases the life span. Accordingly, life
span is significantly shorter in Klotho-deficient mice (klotho(-/-)) than in
their wild-type littermates (klotho(+/+)). The pleotropic effects of Klotho
include inhibition of 1,25-dihydroxyvitamin D(3)(1,25(OH)(2)D(3)) formation.
Vitamin D-deficient diet reverses the shortening of life span in klotho(-/-)
mice. In a variety of cells, 1,25(OH)(2)D(3) stimulates Ca(2+) entry. In
erythrocytes, increased Ca(2+) entry stimulates suicidal erythrocyte death,
which is characterized by cell shrinkage and phosphatidylserine exposure at the
erythrocyte surface. The present study explored the putative impact of Klotho on
eryptosis. According to Fluo3 fluorescence, cytosolic Ca(2+) concentration was
significantly larger in klotho(-/-) erythrocytes as compared to klotho(+/+)
erythrocytes. According to annexin V-binding, phosphatidylserine exposure was
significantly enhanced, and according to forward scatter, cell volume
significantly decreased in klotho(-/-) erythrocytes as compared to klotho(+/+)
erythrocytes. Energy depletion (13 h glucose depletion) and oxidative stress (35
min 1 mM tert-butyl-hydroxyl-peroxide [tert-BOOH]) increased phosphatidylserine
exposure to values again significantly larger in klotho(-/-) erythrocytes as
compared to klotho(+/+) erythrocytes. Reticulocyte number was significantly
increased in klotho (-/-) mice, pointing to enhanced erythrocyte turnover.
Vitamin D-deficient diet reversed the enhanced Ca(2+) entry and annexin
V-binding of klotho(-/-) erythrocytes. The present observations reveal a novel
function of Klotho, i.e., the at least partially vitamin D-dependent regulation
of cytosolic Ca(2+) activity in and suicidal death of erythrocytes. Serum phosphate concentration value is controlled by the kidney, which adapts
phosphate reabsorption in the proximal tubule to the needs of the body and
regulates intestine absorption of phosphate and calcium through calcitriol
synthesis. Fibroblast Growth Factor 23 (FGF23) is a hormone that controls
sodium-phosphate transporter and 1-alpha 25(OH) vitamin D hydroxylase expression
in the renal proximal tubule. FGF23 is synthesized by bone cells in response to
an increase in serum phosphate or calcitriol concentrations. The binding of
FGF23 to a FGF receptor requires a protein named Klotho that is expressed in the
kidney in the distal but not in the proximal tubule. The mechanism by which
FGF23 controls proximal tubule function remains to be established. The
alteration of the FGF23-Klotho axis in animal models and various human disorders
is associated with abnormal control of body phosphate content confirming the
major role played by these protein in phosphate homeostasis. This review details
FGF23 and Klotho functions in normal conditions and in genetics or acquired
disorders. The klotho gene (KL) was identified first as a putative aging-suppressor gene
that extended life span when overexpressed and accelerated aging-like phenotypes
when disrupted in mice. It encodes a single-pass transmembrane protein and is
expressed predomitly in kidney, where it functions as an obligate coreceptor
for fibroblast growth factor 23 (FGF-23). FGF-23 is a bone-derived hormone that
suppresses phosphate reabsorption and 1,25 dihydroxyvitamin D(3) (vitamin D)
synthesis in the kidney. Klotho also is expressed in the parathyroid gland,
where FGF-23 decreases parathyroid hormone expression and secretion, further
suppressing vitamin D synthesis in kidney. Thus, FGF-23 functions as a
phosphaturic hormone and a counter-regulatory hormone for vitamin D, thereby
inducing negative phosphate balance. Mice lacking either FGF-23 or Klotho show
hyperphosphatemia in addition to developing multiple aging-like phenotypes,
which can be rescued by resolving phosphate retention. These findings have
unveiled an unexpected link between aging and phosphate. In patients with
chronic kidney disease (CKD), phosphate retention is seen universally and has
been associated with increased mortality risk. Patients with CKD have high serum
FGF-23 levels with decreased klotho expression in the kidney and parathyroid,
rendering FGF-23 and Klotho as potential biomarkers and therapeutic targets for
CKD. The Klotho protein not only serves as a coreceptor for FGF-23, but also
functions as a humoral factor. Klotho's extracellular domain is released into
blood and urine by ectodomain shedding and exerts various functions
independently of FGF-23, including regulation of multiple ion channels and
transporters. Decreased urinary Klotho protein level has been identified as one
of the earliest biomarkers of CKD progression. This review focuses on the
current understanding of Klotho protein function, with emphasis on its potential
involvement in the pathophysiologic process of CKD. The Klotho gene has been identified as an aging suppressor gene that encodes a
transmembrane protein, which is expressed primarily in renal tubules. There are
2 forms of Klotho, membrane and secreted. However, there is a paucity of data on
levels of soluble Klotho in diseases like diabetes and kidney disease. We
validated an enzyme-linked immunosorbent assay for Klotho and quantitated Klotho
levels separately in patients with diabetes and also in patients with chronic
kidney disease (CKD). The Klotho assay showed good precision and was linear down
to 19 ng/mL. There were no significant effects on Klotho levels with the
addition of common interferents such as ascorbate, triglycerides, or hemolysis;
only bilirubin (250 mg/L) significantly reduced Klotho levels (P < .05). There
was a significant reduction in Klotho levels in samples with glycated hemoglobin
(HbA(1c)) levels of 6.5% or more compared with control samples (HbA(1c) < 6.5%;
P < .001). We also documented significantly higher levels of Klotho with CKD.
Thus, we validated an assay for Klotho and made the novel observation that
levels are decreased in diabetes and increased in CKD. BACKGROUND: Klotho, a single-pass transmembrane protein primarily expressed in
the kidneys, parathyroid glands, and choroid plexus of the brain, has a short
cytoplasmic tail and a long extracellular domain, which can be cleaved and
released as a soluble form. However, information regarding the origins and
kinetics of soluble serum Klotho remains poorly understood. We evaluated serial
changes in serum Klotho levels among living donors before and after
retroperitoneoscopic nephrectomy as well as in their renal transplant
recipients.
METHODS: The levels of soluble Klotho in serum obtained from 10 living donors
and their renal transplant recipients were determined using a sandwich
enzyme-linked immunosorbent assay system.
RESULTS: Serum soluble Klotho was detectable in all subjects. The baseline serum
Klotho concentrations in the living donors ranged from 726.4 to 1417.1 pg/mL
(median, 909.8 pg/mL; interquartile ranges [IR], 754.8-1132.4), whereas that in
the concomitant renal transplant recipients ranged from 397.5 to 1047.2 pg/mL
(median, 613.0 pg/mL; IR, 445.9-750.8; P = .003). The levels of soluble serum
Klotho measured 5 days after retroperitoneoscopic nephrectomy (median, 619.0
pg/mL; IR, 544.6-688.5; P = .001) were significantly lower than the baseline
values. Among the renal transplant recipients, no significant changes in serum
Klotho levels were observed during the observation period.
CONCLUSION: Our data regarding soluble serum Klotho levels obtained from living
donors support the idea that the kidneys are a major source of soluble serum
Klotho in human subjects without a deterioration of renal function. In
recipients, concomitant acute kidney injuries and immunosuppressive protocols
might modulate the release of soluble Klotho from the grafts into the
circulation. AIM: Active vitamin D (1,25-dihydroxyvitamin D₃), PTH, fibroblast growth
factor-23 (FGF-23) and Klotho protein are key regulators of phosphate
metabolism. Hyperphosphatemia and increased FGF-23 level in patients with
end-stage renal disease are associated with increased morbidity and mortality.
The relationships among key regulators of phosphate metabolism are still being
investigated. FGF-23, the humoral factor involved in phosphate metabolism, is
strongly associated with serum phosphorus level. Klotho, a transmembrane protein
expressed primarily in renal tubules, functions as an obligatory co-receptor for
FGF-23. The soluble form of Klotho, produced by the shedding of the
transmembrane protein, is detectable in body fluids. The purpose of the study
was to assess if serum soluble alpha-Klotho level was related to phosphate
metabolism parameters and residual renal function (RRF) in incident peritoneal
dialysis (PD) patients.
METHODS: Thirty-five clinically stable patients 4 to 6 weeks after the onset of
PD were included in the study. For each patient, clinical and laboratory data
were reviewed. Serum phosphorus concentration, urinary and peritoneal phosphate
clearance, serum FGF-23 and soluble Klotho protein concentrations were
determined.
RESULTS: Serum soluble alpha-Klotho was strongly negatively correlated with
24-hour diuresis (Rs = -0.55, p = 0.004) and renal phosphate clearance (Rs =
-0.40, p = 0.049), but not with RRF.
CONCLUSIONS: Serum soluble Klotho protein concentration is inversely related to
residual diuresis and renal phosphate clearance in incident PD patients. Klotho is a recently discovered anti-aging gene and is primarily expressed in
kidneys. In humans, the klotho level decreases with age whereas the prevalence
of chronic kidney disease (CKD) increases with age. Diabetic nephropathy is the
most common form of CKD, which leads to end-stage renal disease. A decrease in
klotho has been found in kidneys of patients with diabetic nephropathy. The
purpose of this study is to assess whether klotho gene deficiency affects early
diabetic nephropathy in a mouse of model of type 1 diabetes induced by
streptozotocin (STZ). Male KL(+/-) mutant and wild-type mice (6-8 weeks) were
injected with multiple low doses of STZ. Renal functions and renal blood flow
were assessed. Kidneys were collected for histological examination and molecular
assays of TGFβ1 and mammalian targets of rapamycin (mTOR) signaling. Klotho
deficiency in KL(+/-) mutant mice exacerbated STZ-induced increases in urine
albumin, blood urea nitrogen, expansion of mesangial matrix in renal glomeruli,
and kidney hypertrophy, suggesting a protective role of klotho in kidney
function and structure. Klotho deficiency did not affect renal blood flow.
Notably, klotho deficiency significantly increased phosphorylation of Smad2,
indicating enhanced TGFβ1 signaling in kidneys. Klotho deficiency also increased
phosphorylation of mTOR and S6 (a downstream effector of mTOR), indicating
enhanced mTOR signaling in kidneys of early diabetic mice. Thus, klotho gene
deficiency may make kidneys more susceptible to diabetic injury. Klotho gene
deficiency exacerbated early diabetic nephropathy via enhancing both TGFβ1 and
mTOR signaling in kidneys. Klotho is a transmembrane protein expressed primarily in kidney, parathyroid
gland, and choroid plexus. The extracellular domain could be cleaved off and
released into the systemic circulation. Klotho is in part effective as
β-glucuronidase regulating protein stability in the cell membrane. Klotho is a
major determit of aging and life span.Overexpression of Klotho increases and
Klotho deficiency decreases life span. Klotho deficiency may further result in
hearing loss and cardiac arrhythmia. The present study explored whether Klotho
modifies activity and protein abundance of KCNQ1/KCNE1, a K(+) channel required
for proper hearing and cardiac repolarization. To this end, cRNA encoding
KCNQ1/KCNE1 was injected in Xenopus oocytes with or without additional injection
of cRNA encoding Klotho. KCNQ1/KCNE1 expressing oocytes were treated with human
recombit Klotho protein (30 ng/mL) for 24 h. Moreover, oocytes which express
both KCNQ1/KCNE1 and Klotho were treated with 10 μM DSA L (D-saccharic
acid-1,4-lactone), a β-glucuronidase inhibitor. The KCNQ1/KCNE1
depolarization-induced current (I(Ks)) was determined utilizing dual electrode
voltage clamp, while KCNQ1/KCNE1 protein abundance in the cell membrane was
visualized utilizing specific antibody binding and quantified by
chemiluminescence. KCNQ1/KCNE1 channel activity and KCNQ1/KCNE1 protein
abundance were upregulated by coexpression of Klotho. The effect was mimicked by
treatment with human recombit Klotho protein (30 ng/mL) and inhibited by DSA
L (10 μM). In conclusion, Klotho upregulates KCNQ1/KCNE1 channel activity by
“mainly” enhancing channel protein abundance in the plasma cell membrane, an
effect at least partially mediated through the β-glucuronidase activity of
Klotho protein. The antiaging protein of Klotho is a transmembrane protein mainly expressed in
the kidney, parathyroid glands and choroid plexus of the brain. The Klotho
protein exists in two forms, a full-length membrane form and a soluble secreted
form. The extracellular domain of Klotho can be enzymatically cleaved off and
released into the systemic circulation where it acts as β-glucuronidase and a
hormone. Soluble Klotho can be found in the blood, cerebrospinal fluid, and the
urine of mammals. Klotho deficiency results in early appearance of multiple
age-related disorders and premature death, whereas overexpression of Klotho
exerts the opposite effect. Klotho may influence cellular transport processes
across the cell membrane by inhibiting calcitriol (1,25(OH) (2)D(3)), formation
or by directly affecting transporter proteins, including ion channels, carriers
and pumps. Accordingly, Klotho protein is a powerful regulator of transport
mechanisms across the cell membrane. Klotho regulates diverse calcium and
potassium ion channels, as well as several carriers including the Na(+)-coupled
excitatory amino acid transporters EAAT3 and EAAT4, the Na(+)-coupled phosphate
cotransporters, NaPi-IIa and NaPi-IIb, and a Na(+)/K(+)-ATPase. All those
cellular transport regulations contribute in the aging suppressor role of
Klotho. Future studies will help to determine if the Klotho protein regulates
cell-surface expression of other transport proteins and is affecting underlying
mechanisms. Klotho is a membrane-bound protein predomitly expressed in the kidney, where
it acts as a permissive co-receptor for Fibroblast Growth Factor 23. In its shed
form, Klotho exerts anti-fibrotic effects in several tissues. Klotho-deficient
mice spontaneously develop fibrosis and Klotho deficiency exacerbates the
disease progression in fibrotic animal models. Furthermore, Klotho
overexpression or supplementation protects against fibrosis in various models of
renal and cardiac fibrotic disease. These effects are mediated at least
partially by the direct inhibitory effects of soluble Klotho on TGFβ1 signaling,
Wnt signaling, and FGF2 signaling. Soluble Klotho, as present in the
circulation, appears to be the primary mediator of anti-fibrotic effects.
Similarly, through inhibition of the TGFβ1, Wnt, FGF2, and IGF1 signaling
pathways, Klotho also inhibits tumorigenesis. The Klotho promoter gene is
generally hypermethylated in cancer, and overexpression or supplementation of
Klotho has been found to inhibit tumor growth in various animal models. This
review focuses on the protective effects of soluble Klotho in inhibiting renal
fibrosis and fibrosis in distant organs secondary to renal Klotho deficiency. We
also discuss the structure-function relationships of Klotho domains and
biological effects in the context of potential targeted treatment strategies. BACKGROUND: The hormone klotho, encoded by the gene klotho, is primarily
expressed in the kidney and choroid plexus of the brain. Higher klotho
concentrations have been linked to better physical performance; however, it is
unknown whether klotho relates to frailty status in older adults.
METHODS: Plasma klotho was measured in 774 participants aged ≥65 years enrolled
in InCHIANTI, a prospective cohort study comprising Italian adults. Frailty
status was assessed at 3 and 6 years after enrollment. Frailty was defined as
presence of at least three out of five criteria of unintentional weight loss,
exhaustion, sedentariness, muscle weakness, and slow walking speed; prefrailty
was defined as presence of one or two criteria; and robustness was defined as
zero criteria. We assessed whether plasma klotho concentrations measured at the
3-year visit related to frailty.
RESULTS: Each additional natural logarithm of klotho (pg/mL) was associated with
lower odds of frailty versus robustness after adjustment for covariates (odds
ratio [OR] 0.46; 95% confidence interval 0.21, 0.98; p-value = .045). Higher
klotho was particularly associated with lower odds of exhaustion (OR 0.57; 95%
CI 0.36, 0.89; p-value = .014). Participants with higher klotho also had lower
estimated odds of weight loss and weakness, but these findings were not
statistically significant.
CONCLUSIONS: Higher plasma klotho concentrations were associated with lower
likelihoods of frailty and particularly exhaustion. Future studies should
investigate modifiable mechanisms through which klotho may affect the frailty
syndrome. Aerobic exercise induces many adaptive changes in the whole body and improves
metabolic characteristics. Klotho, an anti-aging gene, is mainly expressed in
the brain and kidney. The roles of Klotho in the brain and kidney during aerobic
exercise remain largely unknown. The present study aimed to determine whether
aerobic exercise could influence the expression of Klotho, decrease reactive
oxygen species (ROS) and prolong life span. Sprague Dawley rats were exercised
on a motor treadmill. Klotho mRNA and protein expression levels in rat brain and
kidney tissues were examined using reverse transcription-quantitative polymerase
chain reaction and western blotting, respectively. ROS production was detected
following intermittent aerobic exercise (IAE) or continuous aerobic exercise
(CAE). Kaplan-Meier curve analysis demonstrated that aerobic exercise
significantly improved rat survival (P<0.001). The ROS levels in rat brain and
kidney tissues were decreased in the aerobic exercise groups compared with the
control group (P<0.05). In addition, Klotho mRNA and protein expression levels
were increased significantly following aerobic exercise compared with controls
(P<0.05). There was no significant difference between the IAE and CAE groups in
any experiments (P>0.05). These results suggest that aerobic exercise-stimulated
Klotho upregulation extends the life span by attenuating the excess production
of ROS in the brain and kidney. As Klotho exhibits a potential anti-aging
effect, promoting Klotho expression through aerobic exercise may be a novel
approach for the prevention and treatment of aging and aging-related diseases. Klotho is a protein primarily expressed in renal tubular epithelial cells.
Studies have suggested that Klotho is an antiaging protein that reduces renal
fibrosis after acute kidney injury (AKI) and inhibits stem cell senescence. Bone
marrow mesenchymal stem cells (BMSCs) have consistent proliferation ability and
multidirectional differentiation ability and have been used to treat tissue
injury. Thus, we hypothesized that Klotho expressed in BMSCs could increase the
renal protective effects of BMSCs. To verify the hypothesis, we isolated BMSCs
from C57BL/6 mice, transfected them with Klotho-GFP-adenovirus and investigated
the change in BMSC proliferation. We then transplanted Klotho-GFP-BMSCs into
mice with AKI and investigated the therapeutic effect compared with that of
sham-treated mice and GFP-BMSC-transplanted mice. Kidney fibrosis after
ischemia/reperfusion injury (IRI) was relieved by BMSC transplantation, and the
antifibrotic effect of BMSCs was significantly enhanced by overexpressing the
Klotho gene. Mechanistic studies showed that Klotho increased pluripotency gene
expression in BMSCs. Klotho produced by Klotho-GFP-BMSCs inhibited the
Wnt/β-catenin pathway in renal tubular epithelial cells (TECs). Klotho-GFP-BMSCs
showed increased proliferative ability and more potent immuno-regulation ability
than did GFP-BMSCs. Our findings suggested that Klotho gene-modified BMSCs may
be a better choice for cell therapy after AKI. α-Klotho, a multifunctional protein, has been demonstrated to protect tissues
from injury via anti-oxidation and anti-inflammatory effects. The expression of
α-klotho is regulated by several physiological and pathological factors,
including acute inflammatory stress, oxidative stress, hypertension, and chronic
renal failure. Exhaustive exercise has been reported to result in tissue damage,
which is induced by inflammation, oxidative stress, and energy metabolism
disturbance. However, little is known about the effects of exhaustive exercise
on the expression of α-klotho in various tissues. To determine the effects, the
treadmill exhaustion test in mice was performed and the mice were sacrificed at
different time points following exhaustive exercise. Our results confirmed that
the full-length (130 kDa) and shorter-form (65 kDa) α-klotho were primarily
expressed in the kidneys. Moreover, we found that, except for the kidneys and
brain, other tissues primarily expressed the shorter-form α-klotho, including
liver, which was in contrast to previous reports. Furthermore, the shorter-form
α-klotho was decreased immediately following the acute exhaustive exercise and
was then restored to the pre-exercise level or even higher levels in the next
few days. Our results indicate that α-klotho may play a key role in the body
exhaustion and recovery following exhaustive exercise. Klotho interacts with various membrane proteins such as receptors for
transforming growth factor-β (TGF-β) and insulin-like growth factor (IGF). Renal
expression of klotho is diminished in polycystic kidney disease (PKD). In the
present study, the effects of klotho supplementation on PKD were assessed.
Recombit human klotho protein (10 μg·kg-1·day-1) or a vehicle was
administered daily by subcutaneous injection to 6-wk-old mice with PKD
(DBA/2-pcy). Blood pressure was measured using tail-cuff methods. After 2 mo,
mice were killed, and the kidneys were harvested for analysis. Exogenous klotho
protein supplementation reduced kidney weight, cystic area, systolic blood
pressure, renal angiotensin II levels, and 8-epi-PGF2α excretion (P < 0.05).
Klotho protein supplementation enhanced glomerular filtration rate, renal
expression of superoxide dismutase, and klotho itself (P < 0.05). Klotho
supplementation attenuated renal expressions of TGF-β and collagen type I and
diminished renal abundance of Twist, phosphorylated Akt, and mammalian target of
rapamycin (P < 0.05). Pathological examination revealed that klotho decreased
the fibrosis index and nuclear staining of Smad in PKD kidneys (P < 0.05). Our
data indicate that klotho protein supplementation ameliorates the
renin-angiotensin system, reducing blood pressure in PKD mice. Furthermore, the
present results implicate klotho supplementation in the suppression of
Akt/mammalian target of rapamycin signaling, slowing cystic expansion. Finally,
our findings suggest that klotho protein supplementation attenuated fibrosis at
least partly by inhibiting epithelial mesenchymal transition in PKD. The fortuitously discovered antiaging membrane protein αKlotho (Klotho) is
highly expressed in the kidney, and deletion of the Klotho gene in mice causes a
phenotype strikingly similar to that of chronic kidney disease (CKD). Klotho
functions as a co-receptor for fibroblast growth factor 23 (FGF23) signaling,
whereas its shed extracellular domain, soluble Klotho (sKlotho), carrying
glycosidase activity, is a humoral factor that regulates renal health. Low
sKlotho in CKD is associated with disease progression, and sKlotho
supplementation has emerged as a potential therapeutic strategy for managing
CKD. Here, we explored the structure-function relationship and
post-translational modifications of sKlotho variants to guide the future design
of sKlotho-based therapeutics. Chinese hamster ovary (CHO)- and human embryonic
kidney (HEK)-derived WT sKlotho proteins had varied activities in FGF23
co-receptor and β-glucuronidase assays in vitro and distinct properties in vivo
Sialidase treatment of heavily sialylated CHO-sKlotho increased its co-receptor
activity 3-fold, yet it remained less active than hyposialylated HEK-sKlotho. MS
and glycopeptide-mapping analyses revealed that HEK-sKlotho is uniquely modified
with an unusual N-glycan structure consisting of N,N'-di-N-acetyllactose diamine
at multiple N-linked sites, one of which at Asn-126 was adjacent to a putative
GalNAc transfer motif. Site-directed mutagenesis and structural modeling
analyses directly implicated N-glycans in Klotho's protein folding and function.
Moreover, the introduction of two catalytic glutamate residues conserved across
glycosidases into sKlotho enhanced its glucuronidase activity but decreased its
FGF23 co-receptor activity, suggesting that these two functions might be
structurally divergent. These findings open up opportunities for rational
engineering of pharmacologically enhanced sKlotho therapeutics for managing
kidney disease. NEW FINDINGS: What is the central question of this study? Can short-term
high-intensity interval training (HIIT) contribute to the reduction of
ischaemia-reperfusion (IR) injury by enhancing the levels of Klotho and its
related axes, including myocardial TRPC6 expression, and antioxidant defence as
novel possible mechanisms of exercise-induced cadioprotection (EICP) against IR
injury? What is the main finding and its importance? The increase of plasma and
myocardial levels of Klotho as a result of preconditioning with HIIT and
prevention of a significant reduction of Klotho during IR injury can promote
cardioprotection and reduce damage by attenuating myocardial TRPC6 expression
and increasing antioxidant defence. The present findings may provide a new
mechanism in EICP and IR injury, and provide the knowledge to develop preventive
and therapeutic approaches.
ABSTRACT: Cardiovascular disease, especially coronary artery disease, remains a
major cause of morbidity and mortality in the world, and ischaemia-reperfusion
(IR) insult is the main pathological cause leading to death. Exercise training
is associated with a reduced risk of cardiovascular disease and the development
of cardioprotection against IR injury. Therefore, the purpose of this study was
to investigate the effect of preconditioning with high-intensity interval
training (HIIT) on myocardial and plasma levels of Klotho and its related axes
as novel mechanisms of exercise-induced cardioprotection against IR injury.
Seventy male Wistar rats were randomly divided into five groups of control,
HIIT, sham, IR and HIIT group that underwent IR injury (H-IR). The training
group performed five sessions of HIIT on the treadmill. The cardiac IR injury
was induced by ligation of the left anterior descending coronary artery for
30 min followed by 24 h reperfusion. Infarct size and histopathological
assessment of cardiac tissues were determined through Evans
Blue-triphenyltetrazolium chloride and haematoxylin-eosin staining,
respectively. We investigated lipid peroxidation and markers of cardiac injury,
antioxidant enzymes and the plasma levels of Klotho using enzyme-linked
immunosorbent assays. Also, myocardial levels of Klotho and TRPC6 expression
were determined by western blot assays. The results demonstrated a significant
increase in myocardial and plasma levels of Klotho following HIIT and a
significant decrease during IR injury. Myocardial TRPC6 channel expression
increased following IR. HIIT also prevented a significant reduction of Klotho
during IR and consequently reduced the expression of the TRPC6 channel in the
H-IR group compared with the IR group. Furthermore, HIIT decreased the infarct
size, cardiac injury, lipid peroxidation, lactate dehydrogenase, creatine kinase
myocardial band and cardiac troponin-I, and improved total antioxidant capacity
and catalase, superoxide dismutase and glutathione peroxidase activities
following IR injury. The findings of the present study suggest that HIIT
improves cardioprotection against IR injury and reduces cardiac damages through
an increase in myocardial and plasma levels of Klotho and its related axes
(TRPC6 and antioxidant defence). These findings can help to develop preventive
and therapeutic approaches. Increased oxidative stress and inflammation play an important role in the
pathogenesis of diabetic cataract. Klotho, known as an anti-ageing protein, has
antioxidative and anti-inflammatory properties. Klotho is expressed in limited
tissues including the lens. Here we examined whether klotho expression is
decreased in diabetic lens and, if so, whether klotho treatment can prevent
diabetic cataract formation. Streptozotocin (STZ)-induced diabetic rats and
age-matched control rats were treated with vehicle or klotho protein, starting
at 1 week after STZ injection. Twelve weeks after treatment, cataract formation
was observed in diabetic rats but not control rats. Cataract formation and
scores were significantly less in klotho-treated diabetic rats than
vehicle-treated diabetic rats. Levels of klotho in plasma, aqueous humor and
lens were significantly decreased in vehicle-treated diabetic rats, compared
with control rats, but were restored in klotho-treated diabetic rats.
Additionally, vehicle-treated diabetic rats had increased oxidative stress and
inflammation in the lens, which were associated with decreased antioxidant
transcriptional master regulator Nrf2 activity and increased transcription
factor NF-κB activity. All of these findings were ameliorated in klotho-treated
diabetic rats. Notably, klotho treatment did not alter blood glucose in diabetic
rats. These results indicate that klotho reduction may increase susceptibility
of the lens to oxidative and inflammatory insults, promoting cataract formation
under diabetic conditions. Klotho treatment can ameliorate the onset and
progression of diabetic cataract via enhancing Nrf2-mediated antioxidant defense
and suppressing NF-κB-mediated inflammatory responses. Klotho in the lens may be
a novel therapeutic target for prevention of cataract formation in diabetes. While radiation nephropathy is a major problem associated with radiotherapy, the
exact mechanisms underlying its pathogenesis and the mediators involved in
kidney deterioration remain to be elucidated. In view of the finding that
senescence is typically increased post‑irradiation, the present study examined
whether ionizing radiation may cause kidney injury by enhancing premature
senescence. The present study explored the relevance of the aging suppressor,
Klotho, which has anti‑aging activity and is highly expressed in murine renal
cells/kidney tissues, under irradiation conditions. Firstly, the effects of
radiation on mouse inner medullary collecting duct‑3 (mIMCD‑3) cells and kidney
tissues of mice were assessed. Subsequently, the mRNA expression levels of
Klotho, TNF‑α and ADAM metallopeptidase domain (ADAM)9/10/17 were analyzed by
reverse transcription‑quantitative PCR following exposure to radiation. In
addition, the levels of these proteins were measured by western blotting or
ELISA. The results revealed that irradiation of mIMCD‑3 cells clearly triggered
cellular senescence. Notably, Klotho gene expression was considerably decreased
in radiation‑exposed mIMCD‑3 cells and in the kidney tissues of irradiated
BALB/c mice, and the corresponding translated protein was consistently expressed
following radiation exposure. Moreover, expression of TNF‑α, a negative
regulator of Klotho, was significantly increased, whereas ADAM9/10/17, an
ectodomain shedding enzyme of Klotho, was decreased in irradiated mIMCD‑3 cells
and in the kidney tissues of BALB/c mice. Collectively, these data suggested
that TNF‑α‑mediated inhibition of Klotho expression and blockage of soluble
Klotho formation via decreased ADAM expression following irradiation may
contribute to the development of renal dysfunction through acceleration of
radiation‑induced cellular senescence. |
Which particular intersex phenotype is related to steroid reductase? | Steroid reductase mutations are associated with both sex-determining and non-syndromic hypospadias | In the 20 yr since it was established that impairment of dihydrotestosterone
formation is the cause of a rare form of human intersex, a wealth of information
has accumulated about the genetics, endocrinology, and variable phenotypic
manifestations, culminating in the cloning of cDNAs encoding two 5
alpha-reductase genes and documentation that mutations in the steroid 5
alpha-reductase 2 gene are the cause of 5 alpha-reductase deficiency. Perplexing
and difficult problems remain unresolved, e.g. whether the variability in
manifestations is due to variable expressions of steroid 5 alpha-reductase 1 or
to effects of testosterone itself. It is also imperative to establish whether
defects in steroid 5 alpha-reductase 2, perhaps in the heterozygous state, are
responsible for a portion of cases of sporadic hypospadias, to determine whether
5 alpha-reductase plays a role in progesterone action in women, and to elucidate
the relation between androgen action and gender role behavior. Conversion of testosterone (T) to dihydrotestosterone (DHT) in genital tissue is
catalysed by the enzyme 5 alpha-reductase 2, which is encoded by the SRD5A2
gene. The potent androgen DHT is required for full masculinization of the
external genitalia. Mutations of the SRD5A2 gene inhibit enzyme activity,
diminish DHT formation, and hence cause masculinization defects of varying
degree. The classical syndrome, formerly described as pseudovaginal
perineoscrotal hypospadias, is characterized by a predomitly female phenotype
at birth and significant virilization without gynecomastia at puberty. We
investigated nine patients with steroid 5 alpha-reductase 2 deficiency (SRD).
Phenotypes, which were classified according to the severity of the
masculinization defect, varied between completely female (SRD type 5),
predomitly female (SRD type 4), ambiguous (SRD type 3), predomitly male
with micropenis and hypospadias (SRD type 2), and completely male without overt
signs of undermasculinization (SRD type 1). T/DHT-ratios were highly increased (
> 50) in the classical syndrome (SRD type 5), but variable in the less severe
affected patients (SRD types 1-4) (14-35). Mutations in the SRD5A2 gene had been
characterized using PCR-SSCP analysis and direct DNA sequencing. A small
deletion was encountered in two patients, while all other patients had single
base mutations which result in amino acid substitutions. We conclude that
phenotypes may vary widely in patients with SRD5A2 gene mutations spanning the
whole range from completely female to normal male without distinctive clinical
signs of the disease. Hence, steroid 5 alpha-reductase deficiency should be
considered not only in sex reversed patients with female or ambiguous
phenotypes, but also in those with mild symptoms of undermasculinization as
encountered in patients with hypospadias and/or micropenis. A classification
based on the severity of the masculinization defect may be used for correlation
of phenotypes with enzyme activities and genotypes, and for comparisons of
phenotypes between different patients as the basis for clinical decisions to be
made in patients with pseudohermaphroditism due to steroid 5 alpha-reductase 2
deficiency. Between 1986 and 1995, a pedigree of six Arabs with male pseudohermaphroditism
due to 5 alpha reductase-2 deficiency have been identified. All, were raised as
girls since birth. At the time of diagnosis, three were post-pubertal, one
pubertal and two pre-pubertal. The external genitalia of 'pseudo-vaginal
perineoscrotal hypospadias' was identical in these subjects. Although these
individuals were a homogeneous group in terms of their sex of upbringing,
phenotypic appearance, endocrinological profile and socio-cultural background,
the development of the gender identity and role was not uniform in these six
cases. Their psycho-sexual make-up was closely related to the transaction of
their life experiences. These cases provide further insight into the interaction
between various factors involved in the development of gender identity and role
in male pseudohermaphrodites in an Eastern culture. Steroid 5-alpha-reductase 2 deficiency is an autosomal recessive disorder with
clinical spectrum ranges from a male phenotype with hypospadia to a female
phenotype with normal wolffian structures. Over 50 different mutations of SRD5A2
gene has been described in affected patients and several mutations were detected
in specific populations. DNAs of two 46,XY DSD Indonesian siblings, aged 13 and
18 years old, with clinically suspected of 5-alpha-reductase deficiency and
their mother were analysed for molecular defects of SRD5A2 gene. Different from
other reports, in our series three mutations were found in each patient. Two
novel mutations were detected in these patients and their mother, which are
p.Gly34fs and c.699-1G>T. The other mutation detected was c.680G>A or
p.Arg227Gln, which commonly described in Far East Asian population. Whether the
p.Arg227Gln mutation is considered a polymorphism or a mutation in Indonesian
population warrants further study. Hypospadias is rarely reported in dogs. In this study we pre-sent 2 novel cases
of this disorder of sexual development and, in addition, a case of hereditary
sex reversal in a female with an enlarged clitoris. The first case was a male
Moscow watchdog with a normal karyotype (78,XY) and the presence of the SRY
gene. In this dog, perineal hypospadias, bilateral inguinal cryptorchidism and
testes were observed. The second case, representing the Cocker spaniel breed,
had a small penis with a hypospadic orifice of the urethra, bilateral
cryptorchidism, testis and a rudimentary gonad inside an ovarian bursa, a normal
female karyotype (78,XX) and a lack of the SRY gene. This animal was classified
as a compound sex reversal (78,XX, SRY-negative) with the hypospadias syndrome.
The third case was a Cocker spaniel female with an enlarged clitoris and
internally located ovotestes. Cytogenetic and molecular analyses revealed a
normal female karyotype (78,XX) and a lack of the SRY gene, while histology of
the gonads showed an ovotesticular structure. This case was classified as a
typical hereditary sex reversal syndrome (78,XX, SRY-negative). Molecular
studies were focused on coding sequences of the SRY gene (case 1) and 2
candidates for monogenic hypospadias, namely MAMLD1 (mastermind-like domain
containing 1) and SRD5A2 (steroid-5-alpha-reductase, alpha polypeptide 2).
Sequencing of the entire SRY gene, including 5'- and 3'-flanking regions, did
not reveal any mutation. The entire coding sequence of MAMLD1 and SRD5A2 was
analyzed in all the intersexes, as well as in 4 phenotypically normal control
dogs (3 females and 1 male). In MAMLD1 2 SNPs, including 1 missense substitution
in exon 1 (c.128A>G, Asp43Ser), were identified, whereas in SRD5A2 7
polymorphisms, including 1 missense SNP (c.358G>A, Ala120Thr), were found. None
of the identified polymorphisms cosegregated with the intersexual phenotype,
thus, we cannot confirm that hypospadias may be associated with polymorphism in
the coding sequence of the studied genes. Steroid 5-alpha-reductase 2 deficiency is a rare disorder leading to male
pseudohermaphroditism, a condition characterized by incomplete differentiation
of male genitalia in 46,XY patients. Here, we report a case of a 21-year-old
woman from Ardabil who presented with primary amenorrhea, ambiguous genitalia,
and lack of breast development. All of the serum hormone profiles were normal
except for raised serum total testosterone. Testosterone to DHT ratio (T/DHT)
was elevated before (15.72) and further increased after hCG stimulation (32.46).
A chromosomal study revealed a 46,XY karyotype. A bilateral gonadectomy,
recessive cliteroplasty, urethroplasty, and vaginoplasty were performed and
hormonal replacement therapy using estrogen was started. In conclusion, the
diagnosis of 5-alpha-reductase 2 deficiency may be suspected in infants with
ambiguous genitalia or in adolescents or young adults with the characteristic
phenotype and serum hormone profiles. Inactivating mutations of the 5α-steroid reductase type-2 (SRD5A2) gene result
in a broad spectrum of masculinization defects, ranging from a male phenotype
with hypospadias to a female phenotype with Wolffian structures. Molecular
studies of the SRD5A2 revealed a new heterozygous gene variant within the coding
region that results in phenotypic expression. A c.92C>T transition changing
serine to phenylalanine at codon 31 of exon 1 (p.Ser31Phe) was identified in a
patient with 46,XY disorder of sexual development who displayed glandular
hypospadias with micropenis and bilateral cryptorchidism. The restoration of the
p.Ser31Phe mutation by site-directed mutagenesis and transient expression assays
using cultured HEK-293 cells showed that this novel substitution does not
abolish but does deregulate the catalytic efficiency of the enzyme. Thus, the
maximum velocity (V max) value was higher for the mutant enzyme (22.5 ± 6.9 nmol
DHT mg protein(-1) h(-1)) than for the wild-type enzyme (9.8 ± 2.0 nmol DHT mg
protein(-1) h(-1)). Increased in vitro activity of the p.Ser31Phe mutant
suggested an activating effect. This case provides evidence that heterozygous
missense mutations in SRD5A2 may induce the abnormal development of male
external genitalia. In 1974, a lack of 5α-dihydrotestosterone (5α-DHT), the most potent androgen
across species except for fish, was shown to be the origin of a type of
pseudohermaphrodism in which boys have female-like external genitalia. This
human intersex condition is linked to a mutation in the steroid-5α-reductase
type 2 (SRD5α2) gene, which usually produces an important enzyme capable of
reducing the Δ4-ene of steroid C-19 and C-21 into a 5α-stereoisomer. Seeing the
potential of SRD5α2 as a target for androgen synthesis, pharmaceutical companies
developed 5α-reductase inhibitors (5ARIs), such as finasteride (FIN) and
dutasteride (DUT) to target SRD5α2 in benign prostatic hyperplasia and
androgenic alopecia. In addition to human treatment, the development of 5ARIs
also enabled further research of SRD5α functions. Therefore, this review details
the morphological, physiological, and molecular effects of the lack of SRD5α
activity induced by both SRD5α mutations and inhibitor exposures across species.
More specifically, data highlights 1) the role of 5α-DHT in the development of
male secondary sexual organs in vertebrates and sex determination in
non-mammalian vertebrates, 2) the role of SRD5α1 in the synthesis of the
neurosteroid allopregolone (ALLO) and 5α-androstane-3α,17β-diol (3α-diol),
which are involved in anxiety and sexual behavior, respectively, and 3) the role
of SRD5α3 in N-glycosylation. This review also features the lesser known
functions of SRD5αs in steroid degradation in the uterus during pregcy and
glucocorticoid clearance in the liver. Additionally, the review describes the
regulation of SRD5αs by the receptors of androgens, progesterone, estrogen, and
thyroid hormones, as well as their differential DNA methylation. Factors known
to be involved in their differential methylation are age, inflammation, and
mental stimulation. Overall, this review helps shed light on the various
essential functions of SRD5αs across species. |
Is atenolol metabolized by CYP2D6? | No, atenolol is metabolized in a CYP2D6-independent manner. | AIMS: The study aimed to investigate the clinical adherence to drug label
recommendations on important drug-drug interactions (DDIs). Dispensing data on
drug combinations involving selective serotonin reuptake inhibitor (SSRI)
antidepressants could help to identify areas for intensified medical education.
METHODS: This was a retrospective, cross-sectional analysis of individual
dispensing data regarding all individuals > or =15 years old in Sweden. The
study analysed the prescribing and dispensing of CYP2D6 drugs (metoprolol,
donepezil, galantamine, codeine, tamoxifen) together with CYP2D6-blocking SSRIs
(paroxetine/fluoxetine) or SSRIs without significant CYP2D6 inhibition
(citalopram/escitalopram/sertraline), and the related prescribing of
CYP2D6-independent comparator drugs (atenolol, rivastigmine, propoxyphene,
anastrozole). Odds were calculated between each CYP2D6 drug and the
corresponding comparator drug in patients on fluoxetine/paroxetine and
citalopram/escitalopram/sertraline, respectively. The odds ratio (OR) was
calculated by dividing the obtained odds in patients on fluoxetine/paroxetine by
the corresponding odds in patients on citalopram/escitalopram/sertraline.
RESULTS: Compared with patients that were dispensed
citalopram/escitalopram/sertraline, patients dispensed fluoxetine/paroxetine had
lower prescribing rates of metoprolol (adjusted OR 0.80; 95% confidence interval
0.76, 0.85), donepezil (0.65; 0.49, 0.86) and galantamine (0.58; 0.41, 0.81). In
contrast, the use of prodrugs codeine (compared woth propoxyphene) or tamoxifen
(compared with anastrozole) was similar among patients on fluoxetine/paroxetine
and citalopram/escitalopram/sertraline (adjusted OR 1.03; 0.94, 1.12 and 1.29;
0.96, 1.73, respectively).
CONCLUSIONS: Clinically important DDIs that are associated with impaired
bioactivation of prodrugs might be more easily neglected in clinical practice
compared with DDIs that cause drug accumulation and symptomatic adverse drug
reactions. |
Describe DeepTRIAGE | DeepTRIAGE (Deep learning for the TRactable Individualised Analysis of Gene Expression) is a novel deep learning architecture which uses an attention mechanism to obtain personalised biomarker scores that describe how important each gene is in predicting the cancer sub-type for each sample. DeepTRIAge simultaneously reveals heterogeneity within the luminal A biomarker score that significantly associate with tumour stage, placing all luminal samples along a continuum of severity. | |
What is Pseudomelanosis duodeni? | Pseudomelanosis duodeni is a rare incidental finding seen on endoscopy and has the characteristic appearance of flat, black-speckled pigmented mucosa that can be associated with gastrointestinal bleeding, hypertension, chronic heart failure, chronic renal failure and consumption of different drugs. | Despite the common practice of upper gastrointestinal endoscopy, the unique
phenomenon of punctate black pigmentation of the duodenal mucosa, now known as
pseudomelanosis duodeni, still remains a rare entity. Four patients with normal
renal function at presentation were found to have this pigment on endoscopy. One
developed renal failure subsequently but the pigmentation persisted and this
observation has not been reported before. Histochemical and ultrastructural
studies were made. It is postulated that the pigment is heterogenous and
represents a form of stored iron. Anti-hypertensive drugs may have a role in the
causation of the pigmentation formation. Pseudomelanosis duodeni, speckled black pigmentation of the duodenal mucosa,
presents a striking appearance at endoscopy. Among the 14 reported cases there
is a predomice of black women greater than 40 years old, but it can occur in
any race and age group. There is no known association with pigmentation
elsewhere in the gastrointestinal tract or with the use of laxatives. However,
most reported patients were hypertensive (many treated with hydralazine and
propranolol) and significant numbers suffered from upper gastrointestinal
bleeding, chronic renal failure, or diabetes mellitus. The pigment is usually
located in mucosal macrophages, in lysosomes. Histochemical studies and electron
probe microanalysis suggest that several pigments may result in this endoscopic
appearance, including lipomelanin, ceroid, iron sulfide, and hemosiderin.
Additional studies, possibly using tissue from surgical resections or autopsies,
are needed to determine the etiology and clinical significance of this
heterogeneous entity. Pseudomelanosis duodeni is an uncommon endoscopic sign characterized by diffuse
small black spots on the first and second portions of the duodenum. It occurs
predomitly in female and elderly patients and is linked to chronic illnesses
and related medications. Between 1988 and 1994, the authors saw eight patients
with pseudomelanosis duodeni. To evaluate the nature of the pigments, special
staining was performed in seven cases. Iron stain was strongly positive in three
cases. Electron microscopy was performed in two cases. This revealed amorphous
bodies within macrophage lysosomes in one case and angular crystals in another
case. These tests suggest that in pseudomelanosis duodeni iron metabolism may be
impaired and iron is pooled within macrophages. Pseudomelanosis duodeni is rarely seen in children. It manifests endoscopically
as peppery speckles in the duodenal mucosa. This pigment corresponds principally
to accumulation of ferrous sulfide in macrophages within the lamina propria. We
report the case of a 16-year-old boy with ectodermal dysplasia who underwent
renal transplantation for vesicoureteral reflux and later developed epigastric
pain. Endoscopic and pathologic findings in the duodenal mucosa were typical of
pseudomelanosis duodeni. A review of the literature reveals shared clinical
features among reported adult and pediatric cases, including chronic renal
failure, use of antihypertensive medication and oral iron supplementation,
and/or presence of gastric hemorrhage. Pseudomelanosis duodeni is a rare entity characterized by dark pigmentation of
duodenal mucosa of uncertain etiology and clinical significance. We report a
case with endoscopic and pathologic correlation. Some aspects about etiology,
clinical and histopathologic characteristics are discussed. Pseudomelanosis duodeni is a rare benign condition. It manifests endoscopically
as discrete, flat, small brown-black spots in the duodenal mucosa. It produces
no symptoms and may be reversible. The cause and natural history of the
pigmentation have not been clarified, although it is associated with a variety
of systemic illnesses and medications. With electron microscopy and
electron-probe energy dispersive X-ray analysis, the pigment corresponds
principally to an accumulation of ferrous sulfide (FeS) in macrophages within
the lamina propria. We report the case of a 56-year-old female patient with a
past history of diabetes mellitus and hypertension. She was admitted because of
nausea, vomiting, and diarrhea and underwent esophagogastroduodenoscopy because
of stool occult blood test results of 3+. Endoscopy revealed diffusely scattered
black spots in the bulb and second portion of the duodenum. Histological
examination showed numerous pigment-laden macrophages in the lamina propria of
mucosal villi. The diagnosis requires further confirmation by electron
microscopy and electron-probe energy dispersive X-ray microanalysis. No special
therapy is indicated for this rare lesion. Pseudomelanosis duodeni is seen endoscopically as dark spots in the duodenal
mucosa and is generally considered to be local deposition of iron from oral iron
intake. However, pseudomelanosis duodeni may be identified histologically even
before it becomes endoscopically evident; iron stainability within the mucosa is
uneven and unpredictable, and multiple clinical conditions other than oral iron
intake may be associated. We reviewed 17 adult patients with histologically
detected pseudomelanosis duodeni, their endoscopic appearances, iron
stainability, and clinical findings including oral iron and drug intake. Only
6/17 (35 %) had endoscopically apparent dark spots. Perl's iron stain was
entirely positive in 18 %, partially positive in 64 %, and negative in 18 % of
cases. History of oral iron was present in 76 % of patients, but other clinical
conditions consistently associated were hypertension in 88 %, end stage renal
disease in 59 %, and diabetes mellitus in 35 % of patients. Pseudomelanosis duodeni (PD) is a rare dark speckled appearance of the duodenum
associated with gastrointestinal bleeding, hypertension, chronic heart failure,
chronic renal failure and consumption of different drugs. We report four cases
of PD associated with chronic renal failure admitted to the gastroenterology
outpatient unit due to epigastric pain, nausea, melena and progressive reduction
of hemoglobin index. Gastroduodenal endoscopy revealed erosions in the esophagus
and stomach, with no active bleeding at the moment. In addition, the duodenal
mucosa presented marked signs of melanosis; later confirmed by histopathological
study. Even though PD is usually regarded as a benign condition, its
pathogenesis and clinical significance is yet to be defined. Pseudomelanosis duodeni is a rare entity characterised by dark pigmented
intracellular granules seen within macrophages that lie within the lamina
propria of the duodenal villi. There is no known treatment, and the clinical
significance and long-term sequelae of this entity are unclear. We present a
case of pseudomelanosis duodeni in a 54-year-old woman who presented with a
1-month history of nausea, vomiting and non-bloody diarrhoea. The medical
history was significant for diabetes mellitus type 2, end-stage renal disease
status postkidney transplant, hypertension, anaemia of chronic disease and
hypothyroidism. A gastroduodenal endoscopy revealed pigmented dark lesions in
the duodenal mucosa. Biopsies from the second part of the duodenum and duodenal
bulb showed pigmented macrophages in the lamina propria. The findings were
consistent with duodenal melanosis. In spite of renal transplant with
normalisation of renal function, the duodenal melanosis persists, which raises
questions on the role of renal impairment in this entity. A 76-year-old female patient with a past medical history of diabetes mellitus,
stage 3 chronic renal failure and iron deficiency anemia was referred for
esophagogastroduodenoscopy (EGD) for evaluation of solid food dysphagia. She had
been on oral therapy with ferrous sulfate for several years. Besides a
Schatzki's ring the EGD revealed a duodenal mucosa with black-speckled
pigmentation. Biopsies were performed and disclosed the deposition of brown
(hemosiderin) pigment within macrophages in the lamina propria of normal villi.
This endoscopic appearance is called pseudomelanosis duodeni (PD). Melanosis of the stomach and duodenum is a rare entity and a striking finding
diagnosed by upper gastrointestinal endoscopy. Here, we describe the case of an
83-year-old female, with a complicated medical history, who was referred to
gastroenterologist to assess bleeding risk. From the endoscopy, it was
determined that she had both melanosis gastri and duodeni. Although both are
rare, gastric melanosis appears to be even more unusual than duodenal melanosis,
with only a few reported cases documented in the literature thus far. Duodenal pseudomelanosis (or pseudomelanosis duodeni) is a rare benign condition
characterized by black-brown speckled pigmentation of the duodenal mucosa.
Collections of pigment-laden macrophages are found in the tips of duodenal
villi. The pigment is thought to be mostly composed of ferrous sulfide.
Histochemichal stains for iron (Perl's prussian blue) or melanin
(Masson-Fontana) may be positive, but are usually negative or unpredictable.
Duodenal pseudomelanosis occurs predomitly in middle-aged to old adults and
more commonly in females. It is associated with chronic renal failure, arterial
hypertension, diabetes mellitus and gastrointestinal bleeding. Medications such
as ferrous sulfate, hydralazine, propranolol, hydrochlorothiazide and furosemide
are thought to play a role as well. We report a case of a 86-year-old female who
presented with a history of watery diarrhea and melena. The patient had a
history of high blood pressure and ischemic stroke episodes. She was on multiple
medication including hidralazine, captopril, hydrochlorthiazide and aspirin. She
was dehydrated, her blood pressure was 96 × 60 mmHg and neurologic examination
showed complete left hemiplegia with central VII nerve palsy. Laboratory tests
showed normal serum electrolytes and renal function. Hemoglobin level was 10.7
g%. An upper endoscopy showed multiple diminutive black spots throughout the
distal duodenal bulb and second portion. Histology showed multiple foci of a
brown-black granular pigment inside macrophages within the tips of the villi
(pseudomelanosis). Stains for iron and melanin were negative. She was treated
with omeprazol, parenteral fluid replacement with saline and partial fasting.
After complete recovery she was discharged for ambulatory follow up. Pseudomelanosis duodeni is a rare incidental finding seen on endoscopy and has
the characteristic appearance of flat, black-speckled pigmented mucosa. We
present the case of an 83-year-old woman who presented with gastrointestinal
bleeding and was found to have pseudomelanosis duodeni. The finding has no
diagnostic or prognostic significance. Therapeutic chelation or endoscopic
follow-up is not recommended. |
Is the glucocorticoid receptor a transcription factor? | Yes,
The glucocorticoid receptor (GR) is a ligand-activated transcription factor that translocates to the nucleus upon hormone stimulation and distributes between the nucleoplasm and membraneless compartments named nuclear foci. | An important site for bovine herpesvirus 1 (BoHV-1) latency is sensory neurons
within trigeminal ganglia (TG). The synthetic corticosteroid dexamethasone
consistently induces BoHV-1 reactivation from latency. Expression of four
Krüppel-like transcription factors (KLF), i.e., KLF4, KLF6, PLZF (promyelocytic
leukemia zinc finger), and KLF15, are induced in TG neurons early during
dexamethasone-induced reactivation. The glucocorticoid receptor (GR) and KLF15
form a feed-forward transcription loop that cooperatively transactivates the
BoHV-1 immediate early transcription unit 1 (IEtu1) promoter that drives bovine
infected cell protein 0 (bICP0) and bICP4 expression. Since the bICP0 gene also
contains a separate early (E) promoter, we tested the hypothesis that GR and KLF
family members transactivate the bICP0 E promoter. GR and KLF4, both pioneer
transcription factors, cooperated to stimulate bICP0 E promoter activity in a
ligand-independent manner in mouse neuroblastoma cells (Neuro-2A). Furthermore,
GR and KLF4 stimulated productive infection. Mutating both half GR binding sites
did not significantly reduce GR- and KLF4-mediated transactivation of the bICP0
E promoter, suggesting that a novel mechanism exists for transactivation. GR and
KLF15 cooperatively stimulated bICP0 activity less efficiently than GR and KL4:
however, KLF6, PLZF, and GR had little effect on the bICP0 E promoter. GR, KLF4,
and KLF15 occupied bICP0 E promoter sequences in transfected Neuro-2A cells. GR
and KLF15, but not KLF4, occupied the bICP0 E promoter at late times during
productive infection of bovine cells. Collectively, these studies suggest that
cooperative transactivation of the bICP0 E promoter by two pioneer transcription
factors (GR and KLF4) correlates with stimulating lytic cycle viral gene
expression following stressful stimuli.IMPORTANCE Bovine herpesvirus 1 (BoHV-1),
an important bovine pathogen, establishes lifelong latency in sensory neurons.
Reactivation from latency is consistently induced by the synthetic
corticosteroid dexamethasone. We predict that increased corticosteroid levels
activate the glucocorticoid receptor (GR). Consequently, viral gene expression
is stimulated by the activated GR. The immediate early transcription unit 1
promoter (IEtu1) drives expression of two viral transcriptional regulatory
proteins, bovine infected cell protein 0 (bICP0) and bICP4. Interestingly, a
separate early promoter also drives bICP0 expression. Two pioneer transcription
factors, GR and Krüppel-like transcription factor 4 (KLF4), cooperatively
transactivate the bICP0 early (E) promoter. GR and KLF15 cooperate to stimulate
bICP0 E promoter activity but significantly less than GR and KLF4. The bICP0 E
promoter contains enhancer-like domains necessary for GR- and KLF4-mediated
transactivation that are distinct from those for GR and KLF15. Stress-induced
pioneer transcription factors are proposed to activate key viral promoters,
including the bICP0 E promoter, during early stages of reactivation from
latency. |
What are the five traits associated with metabolic syndrome? | Metabolic syndrome is the concurrent presentation of multiple cardiovascular risk factors, including obesity, insulin resistance, hyperglycemia, dyslipidemia and hypertension. | BACKGROUND: The close association of type 2 diabetes and atherosclerotic
cardiovascular disease (CVD) suggests that they share a common physiologic
antecedent, postulated to be tissue resistance to insulin. Insulin resistance is
associated with a cluster of risk factors recognized as the metabolic syndrome.
OBJECTIVE: To describe the epidemiology of the insulin resistance syndrome, also
known as the metabolic syndrome.
METHODS: Overall obesity, central obesity, dyslipidemia characterized by
elevated levels of triglycerides and low levels of high-density lipoprotein
cholesterol, hyperglycemia, and hypertension are common traits that, when they
occur together, constitute the metabolic syndrome. The World Health Organization
and the National Cholesterol Education Program Adult Treatment Panel III have
proposed working definitions for the syndrome based on these traits.
Cross-sectional and longitudinal epidemiologic studies provide an emerging
picture of the prevalence and outcomes of the syndrome.
RESULTS: National survey data suggest the metabolic syndrome is very common,
affecting about 24% of US adults who are 20 to 70 years of age and older. The
syndrome is more common in older people and in Mexican Americans. People with
the syndrome are about twice as likely to develop CVD and over 4 times as likely
to develop type 2 diabetes compared with subjects who do not have metabolic
syndrome. While this syndrome may have a genetic basis, environmental factors
are important modifiable risk factors for the condition.
CONCLUSIONS: The metabolic syndrome is very common and will become even more
common as populations age and become more obese. Treatment for component traits
is known to reduce the risk for type 2 diabetes and CVD; whether risk is reduced
by treatment of the syndrome, specifically, remains uncertain. Primary care
physicians must recognize that the co-occurrence of risk factors for type 2
diabetes and CVD represents an extremely adverse metabolic state warranting
aggressive risk factor intervention. BACKGROUND: Insulin resistance, obesity, dyslipidemia, and high blood pressure
characterize the metabolic syndrome. In an effort to explore the utility of
different multivariate methods of data reduction to better understand the
genetic influences on the aggregation of metabolic syndrome phenotypes, we
calculated phenotypic, genetic, and genome-wide LOD score correlation matrices
using five traits (total cholesterol, high density lipoprotein cholesterol,
triglycerides, systolic blood pressure, and body mass index) from the Framingham
Heart Study data set prepared for the Genetic Analysis Workshop 13, clinic
visits 10 and 1 for the original and offspring cohorts, respectively. We next
applied factor analysis to summarize the relationship between these phenotypes.
RESULTS: Factors generated from the genetic correlation matrix explained the
most variation. Factors extracted using the other matrices followed a different
pattern and suggest distinct effects.
CONCLUSIONS: Given these results, different methods of multivariate data
reduction may provide unique clues on the clustering of this complex syndrome. Prevention of the metabolic syndrome and treatment of its main characteristics
are now considered of utmost importance in order to combat the epidemic of type
2 diabetes mellitus and to reduce the increased risk of cardiovascular disease
and all-cause mortality. Insulin resistance/hyperinsulinaemia are consistently
linked with a clustering of multiple clinical and subclinical metabolic risk
factors. It is now widely recognised that obesity (especially abdominal fat
accumulation), hyperglycaemia, dyslipidaemia and hypertension are common
metabolic traits that, concurrently, constitute the distinctive insulin
resistance or metabolic syndrome. Cross-sectional and prospective data provide
an emerging picture of associations of both physical activity habits and
cardiorespiratory fitness with the metabolic syndrome. The metabolic syndrome,
is a disorder that requires aggressive multi-factorial intervention. Recent
treatment guidelines have emphasised the clinical utility of diagnosis and an
important treatment role for 'therapeutic lifestyle change', incorporating
moderate physical activity. Several previous narrative reviews have considered
exercise training as an effective treatment for insulin resistance and other
components of the syndrome. However, the evidence cited has been less consistent
for exercise training effects on several metabolic syndrome variables, unless
combined with appropriate dietary modifications to achieve weight loss. Recently
published randomised controlled trial data concerning the effects of exercise
training on separate metabolic syndrome traits are evaluated within this review.
Novel systematic review and meta-analysis evidence is presented indicating that
supervised, long-term, moderate to moderately vigorous intensity exercise
training, in the absence of therapeutic weight loss, improves the dyslipidaemic
profile by raising high density lipoprotein-cholesterol and lowering
triglycerides in overweight and obese adults with characteristics of the
metabolic syndrome. Lifestyle interventions, including exercise and
dietary-induced weight loss may improve insulin resistance and glucose tolerance
in obesity states and are highly effective in preventing or delaying the onset
of type 2 diabetes in individuals with impaired glucose regulation. Randomised
controlled trial evidence also indicates that exercise training decreases blood
pressure in overweight/obese individuals with high normal blood pressure and
hypertension. These evidence-based findings continue to support recommendations
that supervised or partially supervised exercise training is an important
initial adjunctive step in the treatment of individuals with the metabolic
syndrome. Exercise training should be considered an essential part of
'therapeutic lifestyle change' and may concurrently improve insulin resistance
and the entire cluster of metabolic risk factors. The diagnostic categories of impaired glucose tolerance (IGT) and impaired
fasting glucose (IFG) were stablished in an effort to identify populations at
risk for developing type 2 diabetes mellitus (T2DM). Both IGT and IFG are
associated with increased risk of developing T2DM, but recent analyses found
that the thresholds of risk vary among different populations and an even lower
diagnostic threshold of IFG may be appropriate. IGT has been linked with an
increased risk of cardiovascular events and some analyses have demonstrated an
increased mortality risk compared with patients with normal glucose tolerance.
In contrast, a continuum of increased risk of microvascular manifestations of
T2DM has been demonstrated with IFG but an association of IFG with
cardiovascular events has not been well established. Although both IGT and IFG
are associated with resistance to insulin and increased insulin secretion, they
do not identify the identical patient populations and are not equivalent in
predicting development of T2DM or cardiovascular events. IFG and IGT have been
associated with other features of insulin resistance, including dyslipidaemia,
hypertension, abdominal obesity, microalbuminuria, endothelial dysfunction, and
markers of inflammation and hypercoagulability, traits collectively referred to
as the metabolic syndrome. Analyses of combinations of these components have
also been associated with progression to T2DM, cardiovascular disease and
increased mortality. The foundation of treatment for IGT, IFG, and the metabolic
syndrome is lifestyle modification, including both dietary change and routine
exercise. To date, several clinical trials have found that lifestyle
modification is the most efficacious strategy to prevent progression to T2DM.
Alternative treatments include pharmacotherapy with metformin or acarbose, both
of which have been demonstrated to decrease the development of T2DM. Ongoing
clinical trials are evaluating newer pharmacotherapies, including angiotensin
converting enzyme inhibitors, angiotensin receptor antagonists, metglitinides
and thiazolidinediones, to prevent both T2DM and cardiovascular events. In
combination with lifestyle modification, these therapies offer hope for
effective prevention of T2DM and its consequences in high-risk patients. Metabolic syndrome (MetS) is a common complex trait consisting of the clustering
of abdominal obesity, hypertension, dyslipidemia, and dysglycemia. MetS is found
in about 25% of the population in the United States and is associated with
increased risk for type 2 diabetes and cardiovascular disease. Despite research
into possible genetic influences for MetS, no consistently reproducible genetic
markers have been obtained, partially due to lack of agreement on the definition
of the phenotype. Because phenotypic precision is essential for genomic
interrogation, the evolving discipline of clinical phenomics, which uses
objective and systematic acquisition of phenotypic data (ie, "deep
phenotyping"), may help evaluate the genetic influences of MetS. This article
reviews evidence that MetS has a genetic component and the potential
applicability of clinical phenomics for the genetic evaluation of MetS using the
example of hierarchical cluster analysis of phenotypic components of
lipodystrophy syndromes, which serve as monogenic models of MetS. Obesity is associated with increased susceptibility to dyslipidemia, insulin
resistance, and hypertension, a combination of traits that comprise the
traditional definition of the metabolic syndrome. Recent evidence suggests that
obesity is also associated with the development of nonalcoholic fatty liver
disease (NAFLD). Despite the high prevalence of obesity and its related
conditions, their etiologies and pathophysiology remains unknown. Both genetic
and environmental factors contribute to the development of obesity and NAFLD.
Previous genetic analysis of high-fat, diet-induced obesity in C57BL/6J (B6) and
A/J male mice using a panel of B6-Chr(A/J)/NaJ chromosome substitution strains
(CSSs) demonstrated that 17 CSSs conferred resistance to high-fat, diet-induced
obesity. One of these CSS strains, CSS-17, which is homosomic for A/J-derived
chromosome 17, was analyzed further and found to be resistant to diet-induced
steatosis. In the current study we generated seven congenic strains derived from
CCS-17, fed them either a high-fat, simple-carbohydrate (HFSC) or low-fat,
simple-carbohydrate (LFSC) diet for 16 weeks and then analyzed body weight and
related traits. From this study we identified several quantitative trait loci
(QTLs). On a HFSC diet, Obrq13 protects against diet-induced obesity, steatosis,
and elevated fasting insulin and glucose levels. On the LFSC diet, Obrq13
confers lower hepatic triglycerides, suggesting that this QTL regulates liver
triglycerides regardless of diet. Obrq15 protects against diet-induced obesity
and steatosis on the HFSC diet, and Obrq14 confers increased final body weight
and results in steatosis and insulin resistance on the HFSC diet. In addition,
on the LFSC diet, Obrq 16 confers decreased hepatic triglycerides and Obrq17
confers lower plasma triglycerides on the LFSC diet. These congenic strains
provide mouse models to identify genes and metabolic pathways that are involved
in the development of NAFLD and aspects of diet-induced metabolic syndrome. Moderate-to-high levels of physical activity are established as preventive
factors in metabolic syndrome development. However, there is variability in the
phenotypic expression of metabolic syndrome under distinct physical activity
conditions. In the present study we applied a Genotype X Environment interaction
method to examine the presence of GxEE interaction in the phenotypic expression
of metabolic syndrome. A total of 958 subjects, from 294 families of The
Portuguese Healthy Family study, were included in the analysis. Total daily
energy expenditure was assessed using a 3 day physical activity diary. Six
metabolic syndrome related traits, including waist circumference, systolic blood
pressure, glucose, HDL cholesterol, total cholesterol and triglycerides, were
measured and adjusted for age and sex. GxEE examination was performed on SOLAR
4.3.1. All metabolic syndrome indicators were significantly heritable. The GxEE
interaction model fitted the data better than the polygenic model (p<0.001) for
waist circumference, systolic blood pressure, glucose, total cholesterol and
triglycerides. For waist circumference, glucose, total cholesterol and
triglycerides, the significant GxEE interaction was due to rejection of the
variance homogeneity hypothesis. For waist circumference and glucose, GxEE was
also significant by the rejection of the genetic correlation hypothesis. The
results showed that metabolic syndrome traits expression is significantly
influenced by the interaction established between total daily energy expenditure
and genotypes. Physical activity may be considered an environmental variable
that promotes metabolic differences between individuals that are distinctively
active. Metabolic syndrome (MS), conventionally defined by the presence of at least
three out of five dysmetabolic traits (abdominal obesity, hypertension, low
plasma HDL-cholesterol, high plasma glucose and high triglycerides), has been
associated with an increased risk of several age-related chronic diseases,
including breast cancer (BC). This may have prognostic implications for BC
survivors. 2,092 early stage BC survivors aged 35-70, recruited in eleven
Italian centres 0-5 years after surgical treatment (1.74 years on average), were
followed-up over 2.8 years on average for additional BC-related events,
including BC-specific mortality, distant metastasis, local recurrences and
contralateral BC. At recruitment, 20 % of the patients had MS. Logistic
regression models were carried out to generate OR and 95 % confidence intervals
(CI) for new BC events associated with MS, adjusting for baseline pathological
prognostic factors. New BC events occurred in 164 patients, including 89 distant
metastases. The adjusted ORs for women with MS versus women without any MS
traits were 2.17 (CI 1.31-3.60) overall, and 2.45 (CI 1.24-4.82) for distant
metastasis. The OR of new BC events for women with only one or two MS traits was
1.40 (CI 0.91-2.16). All MS traits were positively associated with new BC
events, and significantly so for low HDL and high triglycerides. MS is an
important prognostic factor in BC. As MS is reversible through lifestyle
changes, interventions to decrease MS traits in BC patients should be
implemented in BC clinics. The metabolic syndrome represents a cluster of closely connected premorbid risk
factors or diseases with visceral obesity type 2 diabetes, hypertension and low
HLD/hypertriglyceridemia as established traits affecting about 20% in the adult
European populations. This syndrome develops on a complex soil with
overnutrition, low physical activity and psychosocial stress. Common
comorbidities are fatty liver, sleep apnoe and endothelial dysfunction with
cardiovascular complications, nephropathy and type 2 diabetes as "end-stage"
diseases. Thus, a rational diagnostic is needed to elucidate the complex cluster
of diseases as basis for an integrated therapy. There is a clear priority for
life style intervention, however, most diseases of the metabolic syndrome need
medical treatment. Medical treatment of single traits has to take into account
possible pleiotropic or adverse effects on the other traits. This paper presents
the pros and cons of major drug intervention for type 2 diabetes, hypertension,
dyslipidemia and hypercoagulation in the context with the metabolic syndrome. In ancient Greek medicine the concept of a distinct syndrome (going together)
was used to label 'a group of signs and symptoms' that occur together and
'characterize a particular abnormality and condition'. The (dys)metabolic
syndrome is a common cluster of five pre-morbid metabolic-vascular risk factors
or diseases associated with increased cardiovascular morbidity, fatty liver
disease and risk of cancer. The risk for major complications such as
cardiovascular diseases, NASH and some cancers develops along a continuum of
risk factors into clinical diseases. Therefore we still include hyperglycemia,
visceral obesity, dyslipidemia and hypertension as diagnostic traits in the
definition according to the term 'deadly quartet'. From the beginning elevated
blood pressure and hyperglycemia were core traits of the metabolic syndrome
associated with endothelial dysfunction and increased risk of cardiovascular
disease. Thus metabolic and vascular abnormalities are in extricable linked.
Therefore it seems reasonable to extend the term to metabolic-vascular syndrome
(MVS) to signal the clinical relevance and related risk of multimorbidity. This
has important implications for integrated diagnostics and therapeutic approach.
According to the definition of a syndrome the rapid global rise in the
prevalence of all traits and comorbidities of the MVS is mainly caused by rapid
changes in life-style and sociocultural transition resp. with over- and
malnutrition, low physical activity and social stress as a common soil. OBJECTIVE: Metabolic syndrome and the presence of metabolic syndrome components
are risk factors for cardiovascular disease (CVD). However, the association
between personality traits and metabolic syndrome remains controversial, and few
studies have been conducted in East Asian populations.
METHODS: We measured personality traits using the Japanese version of the
Eysenck Personality Questionnaire (Revised Short Form) and five metabolic
syndrome components-elevated waist circumference, elevated triglycerides,
reduced high-density lipoprotein cholesterol, elevated blood pressure, and
elevated fasting glucose-in 1322 participants aged 51.1±12.7years old from
Kakegawa city, Japan. Metabolic syndrome score (MS score) was defined as the
number of metabolic syndrome components present, and metabolic syndrome as
having the MS score of 3 or higher. We performed multiple logistic regression
analyses to examine the relationship between personality traits and metabolic
syndrome components and multiple regression analyses to examine the relationship
between personality traits and MS scores adjusted for age, sex, education,
income, smoking status, alcohol use, and family history of CVD and diabetes
mellitus. We also examine the relationship between personality traits and
metabolic syndrome presence by multiple logistic regression analyses.
RESULTS: "Extraversion" scores were higher in those with metabolic syndrome
components (elevated waist circumference: P=0.001; elevated triglycerides:
P=0.01; elevated blood pressure: P=0.004; elevated fasting glucose: P=0.002).
"Extraversion" was associated with the MS score (coefficient=0.12, P=0.0003). No
personality trait was significantly associated with the presence of metabolic
syndrome.
CONCLUSIONS: Higher "extraversion" scores were related to higher MS scores, but
no personality trait was significantly associated with the presence of metabolic
syndrome. Metabolic syndrome is a cluster of the most dangerous heart attack risk factors
(diabetes and raised fasting plasma glucose, abdominal obesity, high cholesterol
and high blood pressure), and has become a major global threat to human health.
A number of studies have demonstrated that hundreds of non-coding RNAs,
including miRNAs and lncRNAs, are involved in metabolic syndrome-related
diseases such as obesity, type 2 diabetes mellitus, hypertension, etc. However,
these research results are distributed in a large number of literature, which is
not conducive to analysis and use. There is an urgent need to integrate these
relationship data between metabolic syndrome and non-coding RNA into a
specialized database. To address this need, we developed a metabolic
syndrome-associated non-coding RNA database (ncRNA2MetS) to curate the
associations between metabolic syndrome and non-coding RNA. Currently,
ncRNA2MetS contains 1,068 associations between five metabolic syndrome traits
and 627 non-coding RNAs (543 miRNAs and 84 lncRNAs) in four species. Each record
in ncRNA2MetS database represents a pair of disease-miRNA (lncRNA) association
consisting of non-coding RNA category, miRNA (lncRNA) name, name of metabolic
syndrome trait, expressive patterns of non-coding RNA, method for validation,
specie involved, a brief introduction to the association, the article
referenced, etc. We also developed a user-friendly website so that users can
easily access and download all data. In short, ncRNA2MetS is a complete and
high-quality data resource for exploring the role of non-coding RNA in the
pathogenesis of metabolic syndrome and seeking new treatment options. The
website is freely available at http://www.biomed-bigdata.com:50020/index.html. BACKGROUND AND AIMS: Metabolic syndrome is the concurrent presentation of
multiple cardiovascular risk factors, including obesity, insulin resistance,
hyperglycemia, dyslipidemia and hypertension. It has been suggested that some of
these risk factors can have detrimental effects on the skeletal muscle while
others can be a direct result of skeletal muscle abnormalities, showing a
two-way directionality in the pathogenesis of the condition. This review aims to
explore this bidirectional correlation by discussing the impact of metabolic
syndrome on skeletal muscle tissue in general and will also discuss ways in
which skeletal muscle alterations may contribute to the pathogenesis of
metabolic syndrome.
METHODS: Literature searches were conducted with key words (e.g. metabolic
syndrome, skeletal muscle, hyperglycemia) using PubMed, EBSCOhost, Science
Direct and Google Scholar. All article types were included in the search.
RESULTS: The pathological mechanisms associated with metabolic syndrome, such as
hyperglycemia and inflammation, have been associated with changes in skeletal
muscle fiber composition, metabolism, insulin sensitivity, mitochondrial
function, and strength. Additionally, some skeletal muscle alterations,
particularly mitochondrial dysfunction and insulin resistance, are suggested to
contribute to the development of metabolic syndrome. For example, the suggested
underlying mechanisms of sarcopenia development are also contributors to
metabolic syndrome pathogenesis.
CONCLUSION: Whilst numerous studies have identified a relationship between
metabolic syndrome and skeletal muscle abnormalities, further investigation into
the underlying mechanisms is needed to elucidate the best prevention and
management strategies for these conditions. |
Which gene is responsible for the Liebenberg syndrome? | Liebenberg syndrome is a genetic disease caused by heterozygous mutations or deletions of the zinc finger E-box-binding homeobox 2 (ZEB2) gene. Patients present with prominent neurological, medical, and behavioral symptoms. | The study of homeotic-transformation mutants in model organisms such as
Drosophila revolutionized the field of developmental biology, but how these
mutants relate to human developmental defects remains to be elucidated. Here, we
show that Liebenberg syndrome, an autosomal-domit upper-limb malformation,
shows features of a homeotic limb transformation in which the arms have acquired
morphological characteristics of a leg. Using high-resolution array comparative
genomic hybridization and paired-end whole-genome sequencing, we identified two
deletions and a translocation 5' of PITX1. The structural changes are likely to
remove active PITX1 forelimb suppressor and/or insulator elements and thereby
move active enhancer elements in the vicinity of the PITX1 regulatory landscape.
We generated transgenic mice in which PITX1 was misexpressed under the control
of a nearby enhancer and were able to recapitulate the Liebenberg phenotype. We report a case of Liebenberg syndrome in a 6-year-old girl, including the
clinical, radiological, angiographic, and operative findings. We note that the
forearm and hand malformations have similarities to leg and foot anatomy. Our
observations may help provide insight into the etiology of this unusual
condition. The Liebenberg syndrome was first described in 1973 in a five- generation
family. A sixth generation was added in 2001, and in 2009 a hitherto unknown
branch of the same family with similar anomalies extended the family tree
significantly. This article describes the clinical findings and illustrates the
abnormalities with radiographs and three-dimensional computed tomography scans.
We discuss the genetic abnormality that causes Liebenberg syndrome, the genomic
rearrangement at the PITX1 locus on chromosome 5.The structural variations seem
to result in an ectopic expression of paired-like homeodomain transcription
factor 1 (PITX1) in the forelimb causing a partial arm-to-leg transformation in
these patients. BACKGROUND: Structural variants (SVs) affecting non-coding cis-regulatory
elements are a common cause of congenital limb malformation. Yet, the functional
interpretation of these non-coding variants remains challenging. The human
Liebenberg syndrome is characterised by a partial transformation of the arms
into legs and has been shown to be caused by SVs at the PITX1 locus leading to
its misregulation in the forelimb by its native enhancer element Pen. This study
aims to elucidate the genetic cause of an unsolved family with a mild form of
Liebenberg syndrome and investigate the role of promoters in long-range gene
regulation.
METHODS: Here, we identify SVs by whole genome sequencing (WGS) and use
CRISPR-Cas9 genome editing in transgenic mice to assign pathogenicity to the
SVs.
RESULTS: In this study, we used WGS in a family with three mildly affected
individuals with Liebenberg syndrome and identified the smallest deletion
described so far including the first non-coding exon of H2AFY. To functionally
characterise the variant, we re-engineered the 8.5 kb deletion using CRISPR-Cas9
technology in the mouse and showed that the promoter of the housekeeping gene
H2afy insulates the Pen enhancer from Pitx1 in forelimbs; its loss leads to
misexpression of Pitx1 by the pan-limb activity of the Pen enhancer causing
Liebenberg syndrome.
CONCLUSION: Our data indicate that housekeeping promoters may titrate
promiscuous enhancer activity to ensure normal morphogenesis. The deletion of
the H2AFY promoter as a cause of Liebenberg syndrome highlights this new
mutational mechanism and its role in congenital disease. |
Which pharmacogenetic test is recommended prior to administering carbamazepine and why? | HLA-B∗15:02 is known as a biomarker for carbamazepine (CBZ) induced Steven-Johnson Syndrome and Toxic Epidermal Necrolysis (SJS/TEN) in some Asian populations. Hence United States Federal Drug Administration (USFDA) recommends HLA-B∗15:02 screening for Asian and other populations with a high prevalence of HLA-B∗15:02, prior to the administration of carbamazepine. | HLA-B∗15:02 is known as a biomarker for carbamazepine (CBZ) induced
Steven-Johnson Syndrome and Toxic Epidermal Necrolysis (SJS/TEN) in some Asian
populations. Hence United States Federal Drug Administration (USFDA) recommends
HLA-B∗15:02 screening for Asian and other populations with a high prevalence of
HLA-B∗15:02, prior to the administration of carbamazepine. This study was
conducted to estimate the prevalence of HLA-B∗15:02 in a cohort of Sri Lankans.
We observed an overall prevalence of 4.3% (4/93) among 93 Sri Lankans comprising
32 Sinhalese, 30 Sri Lankan Tamils and 31 Moors. The allele was detected in 3
[9.3%; 3/32] Sinhalese, 0 [0%; 0/30] Sri Lankan Tamils and in 1 [3%; 1/31] Moor.
The overall prevalence of HLA-B∗15:02 in this population was close to that of
other populations where the USFDA has recommended HLA-B∗15:02 screening. A
larger study is required to confirm these findings, especially among the
Sinhalese where the frequency appears to be high. |
Are there sex differences in oncogenic mutational processes? | Yes. Sex differences have been observed in multiple facets of cancer epidemiology, treatment and biology, and in most cancers outside the sex organs. There are sex-biases in coding and non-coding cancer drivers, mutation prevalence and strikingly, in mutational signatures related to underlying mutational processes. | |
Which drugs are included in the MAID chemotherapy regimen for sarcoma? | MAID chemotherapy regimen for sarcomas include mesna, adriamycin, ifosfamide and dacarbazine. | Although carcinosarcoma occurs in various locations throughout the body, it
rarely originates in the ovary. Chemotherapy has been minimally beneficial. This
case describes a patient with carcinosarcoma of the ovary who responded
minimally to chemotherapy used for epithelial carcinomas but had a complete
response after receiving chemotherapy used for sarcomas. The patient relapsed
within 1 year after receiving cisplatin therapy. She was treated with mesna,
ifosfamide, Adriamycin, and dacarbazine (MAID) chemotherapy and after one cycle
of chemotherapy she had no evidence of tumor. She has received six cycles of
chemotherapy without evidence of progression 13+ months since beginning MAID
therapy. MAID chemotherapy may be useful in the treatment of carcinosarcoma of
the ovary. The mesna, doxorubicin, ifosfamide, dacarbazine regimen produced a 47% response
rate (including 10% complete responses) in 105 eligible adults with advanced
sarcoma. The major dose-limiting toxicity was granulocytopenia. There was one
toxic death from sepsis. Central nervous system and renal toxicity occurred
infrequently, perhaps as a result of the continuous-infusion schedule. This
regimen is being evaluated further in advanced disease, the adjuvant setting,
and in combination with bone marrow colony-stimulating factors. In this phase II trial, 105 eligible patients with no prior chemotherapy and
advanced sarcoma received doxorubicin, ifosfamide, and dacarbazine (DTIC) with
mesna uroprotection (MAID). Starting doses of these drugs were 60, 7,500, and
900 mg/m2 divided over 72 hours by continuous infusion, respectively. Mesna was
given for 84 to 96 hours at 2,500 mg/m2/d. Myelosuppression was dose limiting,
causing the only toxic death (sepsis). Nonhematologic toxicity consisted
predomitly of anorexia and vomiting. Severe mucositis, macroscopic hematuria,
renal tubular acidosis, renal failure, and CNS toxicity occurred in less than 5%
of cycles. No cardiotoxicity was detected. The overall response rate (10%
complete response [CR]) was 47% (95% confidence intervals, 5% to 18% and 37% to
57%, respectively). Most responses (approximately 70%) were observed within two
cycles. Median times to progression were 10 and 9 months, respectively.
Histologic high tumor grade, lesions less than 5 cm, and less than 1 year from
diagnosis to study entry correlated with the probability of response. The median
survival was 16 months. Time from diagnosis to study entry, performance status,
and extent of disease, but not histologic grade, correlated with survival.
Following CR, two patients remain disease-free at 32 and 16 months. Of the 15
additional patients rendered disease-free with surgery, two remain disease-free
at 30 and 18 months with no further therapy. While most relapses occurred in
sites of prior involvement, death from CNS metastases occurred in 11 of the 80
patients with high-grade sarcomas, of whom seven were still responding
systematically (three complete responders). Because of its substantial response
in this phase II trial, the MAID regimen is being compared with doxorubicin and
DTIC alone in advanced sarcomas and to observation in the adjuvant treatment of
high-grade sarcomas in randomized trials. Four patients with metastatic ovarian mixed Müllerian sarcoma (2 homologous, 2
heterologous) were treated with mesna, doxorubicin, ifosfamide, and dacarbazine
(MAID) chemotherapy. Two of four patients had optimal debulking. Three of four
patients responded to chemotherapy, with two complete responses of 34- and
46-month duration. The MAID regimen appears to be active in patients with
ovarian sarcoma. Since dose intensity of doxorubicin is correlated with the clinical response of
patients with soft tissue sarcomas and since doxorubicin dose intensity may be
compromised in combination chemotherapy, we evaluated the use of recombit
granulocytemacrophage colony-stimulating factor (rGM-CSF) to ameliorate
myelosuppression and allow doxorubicin dose escalation in a phase I trial
utilizing the MAID combination [Mesna 2.5 g/m2/day x 4 days, Adriamycin
(doxorubicin) 15 mg/m2/day x 4 days, ifosfamide 2.0 g/m2/day x 3 days,
dacarbazine 250 mg/m2/day x 4 days; to be repeated every 21 days]. Thirteen
patients were treated. The doxorubicin dose for the first 6 patients was at the
standard dose of 15 mg/m2/day x 4 days (level 1), while the doxorubicin dose for
the next 7 patients was escalated by 25% to 18.75 mg/m2/day x 4 days (level 2).
rGM-CSF was given at 5 micrograms/kg/day, days 5-14. All patients experienced
moderate to severe myelosuppression, with all patients at dose level 2 requiring
doxorubicin dose reduction to dose level 1 or lower by their third course of
treatment. rGM-CSF failed to allow sustained escalation of the doxorubicin dose
in the MAID regimen. This study was conducted to determine the maximum tolerated dose of an
intensified MAID (mesna, adriamycin, ifosfamide, dacarbazine) regimen with the
support of lenograstim in patients with advanced soft tissue sarcomas. Following
1 cycle of MAID at the standard dose, four patients were to be treated at each
of five dosage levels: +25%, +45%, +65%, +85%, +100%. Sixteen patients were
treated. Because there were no significant differences in hematologic toxicity
between patients receiving lenograstim 5 or 10 microg/kg/day (levels 1-5 and
1-10), the data were pooled for comparison with level 2. The median duration of
absolute neutrophil count < 0.5 x 10(9)/l was 3 days at level 1 and 7 days at
level 2 (p < 0.01). The median platelet nadir was 25 x 10(9)/l at level 1 and 10
x 10(9)/l at level 2 (p < 0.01). The median duration of toxicity-related
hospitalization was 3.5 days and 11 days at levels 1 and 2, respectively, (p <
0.001). Mucositis > or = grade III occurred after 3/29 cycles at level 1 and
10/15 cycles at level 2 (p < 0.001). After 4 cycles at level 1, 8/8 patients
still had performance status scores < or = 2, and only 4/8 had performance
status scores < or = 2 after the second cycle at level 2. Lenograstim enabled an
increase of 25% of the MAID regimen. At higher dose levels, severe mucositis and
deterioration in performance status were dose limiting. The sarcomatoid variant of renal-cell carcinoma (SRCC), a clinically aggressive
subtype of renal parenchymal tumors, is typically resistant to systemic
treatments and carries a poor prognosis. The authors report a case of a
57-year-old male with advanced SRCC who had a durable complete response after
MAID (mesna, adriamycin, ifosfamide and dacarbazine) chemotherapy, and remains
free of disease four years after completing treatment. To the authors'
knowledge, this is the first report of a remission from MAID chemotherapy in
SRCC. A review of published literature revealed occasional responses after
systemic chemotherapy. Notably, all responses were seen with doxorubicin
containing regimens, suggesting that doxorubicin is a critical component in
chemotherapy regimens for SRCC. BACKGROUND & OBJECTIVES: Chemotherapy combined with other therapeutic modalities
is the main option for advanced and metastatic soft tissue sarcoma(STS). So far
there is no standard regimen for STS yet. Adrimycin, ifosfamide, and dacarbazine
are the most effective agents at present. The purpose of this clinical trial was
to evaluate the efficacy and toxicity of MAID regimen (mesna/ifosfamide +
Adriamycin + dacarbazine) in the treatment of advanced soft tissue sarcoma.
METHODS: Twenty-two patients with advanced STS were treated by MAID(Adriamycin
60 mg/m2, ifosfamide 6,000 mg/m2, and dacarbazine 1,000 mg/m2). These drugs were
administered as continuous intravenous infusion for 72 hours while mesna was
infused continuously for 96 hours.
RESULTS: Partial response rate was 36.4% without complete remission. The
duration of response ranged from 2-10 months with median of 4.6 months. Main
toxicities were myelosuppression, gastrointestinal toxicity and alopecia.
Percentage of leucopenia, nausea/vomiting, and alopecia in WHO grade III and IV
were 63.6%, 27.3%, and 50%, respectively.
CONCLUSIONS: The response rate of MAID for advanced STS was not satisfactory
with evident myelosuppression. Further study on new anti-cancer agents and
regimen are needed. PURPOSE: On the basis of a positive reported single-institution pilot study, the
Radiation Therapy Oncology Group initiated phase II trial 9514 to evaluate its
neoadjuvant regimen in a multi-institutional Intergroup setting.
PATIENTS AND METHODS: Eligibility included a high-grade soft tissue sarcoma > or
= 8 cm in diameter of the extremities and body wall. Patients received three
cycles of neoadjuvant chemotherapy (CT; modified mesna, doxorubicin, ifosfamide,
and dacarbazine [MAID]), interdigitated preoperative radiation therapy (RT; 44
Gy administered in split courses), and three cycles of postoperative CT
(modified MAID).
RESULTS: Sixty-six patients were enrolled, of whom 64 were analyzed.
Seventy-nine percent of patients completed their preoperative CT and 59%
completed all planned CT. Three patients (5%) experienced fatal grade 5
toxicities (myelodysplasias, two patients; infection, one patient). Another 53
patients (83%) experienced grade 4 toxicities; 78% experienced grade 4
hematologic toxicity and 19% experienced grade 4 nonhematologic toxicity.
Sixty-one patients underwent surgery. Fifty-eight of these were R0 resections,
of which five were amputations. There were three R1 resections. The estimated
3-year rate for local-regional failure is 17.6% if amputation is considered a
failure and 10.1% if not. Estimated 3-year rates for disease-free,
distant-disease-free, and overall survival are 56.6%, 64.5%, and 75.1%,
respectively.
CONCLUSION: This combined-modality treatment can be delivered successfully in a
multi-institutional setting. Efficacy results are consistent with previous
single-institution results. BACKGROUND: Metastatic soft tissue sarcoma (STS) prognosis remains poor and few
cytotoxic agents offer proven efficacy. This randomized open phase III study
examines whether high-dose (HD) chemotherapy with peripheral blood stem cells
(PBSCs) could improve overall survival (OS) of chemosensitive patients.
PATIENTS AND METHODS: Advanced STS patients aged 18-65 years received four
courses of standard mesna, adryamycin, ifosfamide and dacarbazine (MAID)
treatment. Chemotherapy-responding patients and patients with at least stable
disease amenable to complete surgical resection were randomized to receive
standard dose (SD) with two successive MAID cycles or HD treatments of one MAID
then MICE intensification: mesna (3.6 g/m(2), day 1-5), ifosfamide (2.5 g/m(2),
day 1-4), carboplatin [area under the curve (AUC) 5/day 2-4] and etoposide (300
mg/m(2), day 1-4) with PBSC reinjection at day 7.
RESULTS: From 2000 to 2008, 207 patients received four cycles of MAID and 87
assessable patients were randomly assigned to receive the following: 46 SD, 41
HD, with 45 and 38 maintained for analyses after secondary centralized
histological review. Futility analyses led to study closure in November 2008.
Three-year OS was 49.4% for the SD group versus 32.7% for HD arm, hazard ratio=
1.26, 95% confidence interval 0.70-2.29; progression-free survival was 32.4% and
14.0%, respectively. HD treatment led to higher grades 3-4 toxicity.
CONCLUSION: This study failed to show an OS advantage for advanced STS patients
treated with dose-intensified chemotherapy with PBSC. BACKGROUND: Good local control of high-grade non-small round cell soft tissue
sarcomas (NSRCSTSs) has been achieved with significant advances in surgical
techniques and radiotherapy. However, the role of chemotherapy remains
controversial. Our aim was to investigate the efficacy, feasibility and adverse
effects of neoadjuvant and adjuvant chemotherapy with modified mesna,
adriamycin, ifosfamide and dacarbazine (MAID) regimen for NSRCSTSs.
METHODS: We conducted a retrospective review of 40 consecutive patients (29 men,
11 women; median age 47 years) with high-grade NSRCSTSs treated in two referral
centers between 2004 and 2009 (median follow-up 38.5 months). Patients with
distant or nodal metastases at diagnosis were excluded. The regimen consisted of
ifosfamide 2,500 mg/m(2)/6 h (days 1-3), mesna 2,500 mg/m(2)/6 h (days 1-3),
tetrahydropyranyl adriamycin 20 mg/m(2)/0.5 h (days 1-3), and dacarbazine 300
mg/m(2)/1 h (days 1-3).
RESULTS: Among the 26 evaluable patients, there were 8 with a partial response,
15 with stable disease, and 3 with progressive disease. Two- and 5-year overall
survival rates were 92 and 86%, respectively, and corresponding disease-free
survival rates were 80 and 77%. All relapses were metastases without local
recurrence. Grade 3-4 neutropenia, anemia and thrombocytopenia were observed in
38, 18, and 21 patients, respectively. No serious infectious complications
occurred due to the administration of granulocyte colony-stimulating factor and
prophylactic antibiotics. No other life-threatening serious adverse events were
observed.
CONCLUSION: The modified MAID regimen achieved a better outcome with less
serious adverse events than previously reported and is a potential option in the
management of NSRCSTSs. Further evaluation with long-term follow-up is required. BACKGROUND: Patients with large, high-grade extremity and truncal soft tissue
sarcomas (STS) are at considerable risk for recurrence. A regimen of
pre-operative chemotherapy consisting of mesna, adriamycin, ifosfamide and
dacarbazine (MAID), interdigitated with radiotherapy (RT), followed by resection
and post-operative chemotherapy with or without RT, has demonstrated high rates
of local and distant control. The goal of this study is to assess outcomes in a
recent cohort of patients treated on this regimen.
METHODS: We retrospectively reviewed records of 66 consecutive patients with STS
of the extremity or trunk who were treated with the aforementioned regimen from
May 2000 to April 2011. Clinicopathologic characteristics and patient outcomes
were analysed.
RESULTS: Sixty-six patients were analysed and were equally divided between grade
2 and 3 tumours. Margins were negative in 57 (89%) patients and positive in
seven (11%) patients. At a median follow-up of 46 months, there were six (9%)
locoregional and 20 (30%) distant recurrences. The locoregional and distant
5-year recurrence-free survival (RFS) rates were 91% and 64%, respectively. The
5-year overall (OS) and disease-specific survival rates were 86% and 89%,
respectively. There were no treatment-related deaths or secondary
myelodysplasias. Thirty-four (52%) patients had grade 3 or 4 acute haematologic
chemotherapy-related toxicity. There were no statistically significant
predictors of OS or RFS.
CONCLUSIONS: For a contemporary cohort of patients with high-risk extremity and
truncal STS, a regimen of neoadjuvant chemoradiotherapy and surgery continues to
result in high rates of survival with tolerable short- and long-term toxicity. Publisher: HINTERGRUND: Für Patienten mit anthrazyklinresistentem metastasierten
Angiosarkom existiert momentan keine Standard-Zweitlinientherapie, und es
besteht Bedarf an neuen effektiven Regimen zur Verbesserung der Ansprechraten.
FALLBERICHT: Wir berichten von einem Fall von primärem, bilateralen Angiosarkom
der Brust bei einer 34-jährigen Patientin mit Lungenmetastasen. Nach 3 Zyklen
des MAID-Regimes (Mesna, Adriamycin, Ifosfamid, Dacarbazin) zeigte die
computertomographische Untersuchung ein Fortschreiten der Erkrankung. Daraufhin
wurde eine Zweitlinienchemotherapie mit dem GVP-Regime (Gemcitabin, Vincristin,
Cisplatin) begonnen. Nach 6 Behandlungszyklen konnte ein komplettes Ansprechen
der Lungenmetastasen verzeichnet werden.
SCHLUSSFOLGERUNG: In Abwesenheit einer effektiven Therapie für Patienten mit
anthrazyklinresistentem metastasierten Angiosarkom der Brust stellt das
GVP-Chemothe-rapieregime eine Behandlungsmöglichkeit dar. OBJECTIVES: Pulmonary pleomorphic carcinoma (PC) is a rare type of lung tumor
with a dismal prognosis. There is no consensus on a chemotherapy regimen for PC,
and conventional platinum-based chemotherapy has been associated with
disappointing response rates and PFS. In searches for a new regimen, the
sarcomatoid (spindle or giant cell) component has been assumed to be susceptible
to chemotherapy used for soft tissue sarcoma.
MATERIALS AND METHODS: The medical records of 17 patients who received mesna,
doxorubicin, ifosfamide, and dacarbazine (MAID) for advanced PC between January
2010 and February 2017 were retrospectively analyzed for clinicopathological
features and outcomes.
RESULTS AND CONCLUSION: The median age was 59 years. Sixteen patients were male,
and only one patient had never smoked. Six patients achieved partial response to
MAID, leading to an objective response rate of 35%. The median PFS was 2.8
months, and the median OS was 8.7 months. Hematologic toxicity-related adverse
events were the most frequent, which comprised grade 3-4 anemia in 35% of
patients, neutropenia in 47%, thrombocytopenia in 24%, and febrile neutropenia
in 29%. No febrile neutropenia was reported in patients who received 5-day
granulocyte-colony stimulating factor (G-CSF) prophylaxis. Most adverse events
resolved without complications, except for one death due to sepsis. MAID is an
effective, and possibly important, regimen for PC. MAID could be more safely
used in clinical practice with appropriate dose modifications and G-CSF primary
prophylaxis according to patients' status. RATIONALE: Ameloblastoma is generally characterized as a benign tumor
originating in odontogenic epithelium. However, few cases of metastatic
maligt ameloblastoma have also been reported. Due to the low incidence of
maligt ameloblastoma, there is no established treatment regimen. To explore
effective treatment for maligt ameloblastoma, we reported this case study.
PATIENTS CONCERNS: This report described a case of a 28-year-old maligt
ameloblastoma female patient with multiple metastasis (brain and lung).
DIAGNOSES: The patient presented ameloblastoma of the left mandible in 2012.
Three years later, local recurrence and brain metastasis was observed during a
follow-up examination. Five years later, maligt ameloblastoma was detected by
imaging and immunohistochemistry in the bilateral multiple pulmonary nodules and
mediastinal lymph nodes.
INTERVENTIONS: The patient was initially treated with tumor resection. Three
years later after local recurrence and brain metastasis, she was accepted the
extensive mandibulectomy supplemented with brain stereotactic body radiotherapy
(SBRT). When diagnosed with pulmonary metastasis, the patient received combined
chemotherapy regimen of MAID (mesna, adriamycin, ifosfamide and dacarbazine) for
6 cycles.
OUTCOMES: The efficacy evaluation was partial remission (PR) after the 6 cycles
of MAID. The last patient follow-up was July 24th 2018, and no evidence of
progression was observed. The progression-free survival (PFS) of the patient was
more than 9 months.
LESSONS: Surgical resection is the optimal treatment for locally recurrent
ameloblastoma. SBRT may be an effective treatment for unresectable
oligometastasis of maligt ameloblastoma. Finally, combined chemotherapy of
MAID showed encouraging effects in the management of metastatic maligt
ameloblastoma. The role of chemotherapy in the treatment of myxofibrosarcoma is unclear. There
are no randomized clinical trials evaluating the therapeutic effect of
chemotherapy on myxofibrosarcoma. We report, to the best of our knowledge, the
first case of myxofibrosarcoma successfully treated with mesna, pirarubicin,
ifosfamide and dacarbazine (modified MAID) regimen. The patient achieved
complete remission evaluated according to Response Evaluation Criteria in Solid
Tumours (RECIST). |
Which is the main ligand for the glucocorticoid receptor? | Glucocorticoids (GC) such as cortisol regulate multiple physiological functions, notably those involved in development, metabolism, inflammatory processes and stress, and exert their effects upon binding to the glucocorticoid receptor (GR, encoded by NR3C1 gene in humans). | Glucocorticoids (GC) such as cortisol regulate multiple physiological functions,
notably those involved in development, metabolism, inflammatory processes and
stress, and exert their effects upon binding to the glucocorticoid receptor (GR,
encoded by NR3C1 gene in humans). GC signaling follows several consecutive steps
leading to target gene transactivation, including ligand binding, nuclear
translocation of ligand-activated GR complexes, DNA binding, and recruitment of
functional transcriptional machinery. Generalized glucocorticoid resistance
syndrome, due to GR loss-of-function mutations, may be related to the impairment
of one of the GC signaling steps. To date, 31 NR3C1 loss-of-function mutations
have been reported in patients presenting with various clinical signs such as
hypertension, adrenal hyperplasia, hirsutism or metabolic disorders associated
with biological hypercortisolism but without Cushing syndrome signs and no
negative regulatory feedback loop on the hypothalamic-pituitary-adrenal axis.
Functional characterization of GR loss-of-function mutations often demonstrates
GR haploinsufficiency and a decrease of GR target gene induction in relevant
cell types. The main signs at presentation are very variable from resistant
hypertension, bilateral adrenal hyperplasia likely related to increased ACTH
levels but not exclusively, hirsutism to isolated renin-angiotensin-aldosterone
system abnormalities in a context of 11βHSD2 deficiency. Some mutated GR
patients are obese or overweight together with a healthier metabolic profile
that remains to be further explored in future studies. Deciphering the molecular
mechanisms altered by GR mutations should enhance our knowledge on GR signaling
and ultimately facilitate management of GC-resistant patients. This review also
focuses on the criteria facilitating identification of novel NR3C1 mutations in
selected patients. Glucocorticoids (GCs) act through the glucocorticoid receptor (GR, also known as
NR3C1) to regulate immunity, energy metabolism and tissue repair. Upon ligand
binding, activated GR mediates cellular effects by regulating gene expression,
but some GR effects can occur rapidly without new transcription. Here, we show
that GCs rapidly inhibit cell migration, in response to both GR agonist and
antagonist ligand binding. The inhibitory effect on migration is prevented by GR
knockdown with siRNA, confirming GR specificity, but not by actinomycin D
treatment, suggesting a non-transcriptional mechanism. We identified a rapid
onset increase in microtubule polymerisation following GC treatment, identifying
cytoskeletal stabilisation as the likely mechanism of action. HDAC6
overexpression, but not knockdown of αTAT1, rescued the GC effect, implicating
HDAC6 as the GR effector. Consistent with this hypothesis, ligand-dependent
cytoplasmic interaction between GR and HDAC6 was demonstrated by quantitative
imaging. Taken together, we propose that activated GR inhibits HDAC6 function,
and thereby increases the stability of the microtubule network to reduce cell
motility. We therefore report a novel, non-transcriptional mechanism whereby GCs
impair cell motility through inhibition of HDAC6 and rapid reorganization of the
cell architecture.This article has an associated First Person interview with the
first author of the paper. A decline in normal physiological functions characterizes the aging process.
While some of these changes are benign, the decrease in the function of the
cardiovascular system that occurs during aging leads to the activation of
pathological processes associated with an increased risk for heart disease and
its complications. Imbalances in endocrine function are also common occurrences
during the aging process. Glucocorticoids are primary stress hormones and are
critical regulators of energy metabolism, inflammation, and cardiac function.
Glucocorticoids exert their actions by binding the glucocorticoid receptor (GR)
and, in some instances, to the mineralocorticoid receptor (MR). GR and MR are
members of the nuclear receptor family of ligand-activated transcription
factors. There is strong evidence that imbalances in GR and MR signaling in the
heart have a causal role in cardiac disease. The extent to which glucocorticoids
play a role in the aging heart, however, remains unclear. This review will
summarize the positive and negative direct and indirect effects of
glucocorticoids on the heart and the latest molecular and physiological evidence
on how alterations in glucocorticoid signaling lead to changes in cardiac
structure and function. We also briefly discuss the effects of other hormones
systems such as estrogens and GH/IGF-1 on different cardiovascular cells during
aging. We will also review the link between imbalances in glucocorticoid levels
and the molecular processes responsible for promoting cardiomyocyte dysfunction
in aging. Finally, we will discuss the potential for selectively manipulating
glucocorticoid signaling in cardiomyocytes, which may represent an improved
therapeutic approach for preventing and treating age-related heart disease. |
Who received the Nobel prize for development of CRISPR? | The 2020 Nobel Prize in Chemistry was awarded to CRISPR-Cas pioneers Emmanuelle Charpentier and Jennifer Doudna. Charpentier and Doudna pioneered the site-specific CRISPR gene-editing technology that has revolutionized cancer research and treatment. | Emmanuelle Charpentier, PhD, and Jennifer Doudna, PhD, who pioneered the
site-specific CRISPR gene-editing technology that has revolutionized cancer
research and treatment, were awarded the 2020 Nobel Prize in Chemistry. Many
CRISPR-based therapies are already in human testing, with gene-edited T cells
for blood cancers and solid tumors leading the way. Conflict of interest statement: Conflict of interest: JB has served on advisory
boards for GenSight Biologics; SparingVision; Akouos, Inc; Life Biosciences; and
Odylia Therapeutics and has consulted for Spark Therapeutics. She served as the
scientific director for clinical trials run by Spark Therapeutics. CRISPR (clustered regularly interspaced short palindromic repeats) is a
prokaryotic immune surveillance system that is used by bacteria to recognize
genetic material of infectious organisms, such as phage viruses. Using
CRISPR-associated (Cas) proteins, this system cleaves foreign nucleic acid into
fragments, thus defending the bacterium against the attacker. The 2020 Nobel
Prize in Chemistry was awarded to CRISPR-Cas pioneers Emmanuelle Charpentier and
Jennifer Doudna, who developed the CRISPR-Cas system to precisely edit genomic
DNA. This technology has exploded at a breathtaking pace and is now used by
almost every molecular biology laboratory around the world in a myriad of
organisms. In this Virtual Issue, the FEBS Journal features articles reviewing
the development of CRISPR/Cas9 technology and its applications to understand the
functions of proteins in vivo. |
How are super enhancers defined? | Super-enhancers are defined as genomic regions spanned by highly conserved non-coding elements (HCNEs), most of which serve as regulatory inputs of one target gene in the region. | Super-enhancers are large clusters of transcriptional enhancers that drive
expression of genes that define cell identity. Improved understanding of the
roles that super-enhancers play in biology would be afforded by knowing the
constellation of factors that constitute these domains and by identifying
super-enhancers across the spectrum of human cell types. We describe here the
population of transcription factors, cofactors, chromatin regulators, and
transcription apparatus occupying super-enhancers in embryonic stem cells and
evidence that super-enhancers are highly transcribed. We produce a catalog of
super-enhancers in a broad range of human cell types and find that
super-enhancers associate with genes that control and define the biology of
these cells. Interestingly, disease-associated variation is especially enriched
in the super-enhancers of disease-relevant cell types. Furthermore, we find that
cancer cells generate super-enhancers at oncogenes and other genes important in
tumor pathogenesis. Thus, super-enhancers play key roles in human cell identity
in health and in disease. Enhancers are critical genomic elements that define cellular and functional
identity through the spatial and temporal regulation of gene expression. Recent
studies suggest that key genes regulating cell type-specific functions reside in
enhancer-dense genomic regions (i.e., super enhancers, stretch enhancers). Here
we report that enhancer RNAs (eRNAs) identified by global nuclear run-on
sequencing are extensively transcribed within super enhancers and are
dynamically regulated in response to cellular signaling. Using Toll-like
receptor 4 (TLR4) signaling in macrophages as a model system, we find that
transcription of super enhancer-associated eRNAs is dynamically induced at most
of the key genes driving innate immunity and inflammation. Unexpectedly, genes
repressed by TLR4 signaling are also associated with super enhancer domains and
accompanied by massive repression of eRNA transcription. Furthermore, we find
each super enhancer acts as a single regulatory unit within which eRNA and genic
transcripts are coordinately regulated. The key regulatory activity of these
domains is further supported by the finding that super enhancer-associated
transcription factor binding is twice as likely to be conserved between human
and mouse than typical enhancer sites. Our study suggests that transcriptional
activities at super enhancers are critical components to understand the dynamic
gene regulatory network. Super-enhancers (SEs) are regions of the genome consisting of clusters of
regulatory elements bound with very high amounts of transcription factors, and
this architecture appears to be the hallmark of genes and noncoding RNAs linked
with cell identity. Recent studies have identified SEs in CD4(+) T cells and
have further linked these regions to single nucleotide polymorphisms (SNPs)
associated with immune-mediated disorders, pointing to an important role for
these structures in the T cell differentiation and function. Here we review the
features that define SEs, and discuss their function within the broader
understanding of the mechanisms that define immune cell identity and function.
We propose that SEs present crucial regulatory hubs, coordinating intrinsic and
extrinsic differentiation signals, and argue that delineating these regions will
provide important insight into the factors and mechanisms that define immune
cell identity. Author information:
(1)Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore,
60 Biopolis Street, Genome #02-01, Singapore 138672, Singapore.
(2)Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, 8
College Road, Singapore 169857, Singapore.
(3)NUS Graduate School for Integrative Sciences and Engineering, National
University of Singapore, 5 Lower Kent Ridge Road, Singapore 119074, Singapore.
(4)Cancer Science Institute of Singapore, National University of Singapore, 14
Medical Drive, #12-01, Singapore 117599, Singapore.
(5)Department of Physiology, Yong Loo Lin School of Medicine, National
University of Singapore, 2 Medical Drive #04-01, Singapore 117597, Singapore.
(6)Department of Human Genetics, Genome Institute of Singapore, 60 Biopolis
Street, Genome #02-01, Singapore 138672, Singapore.
(7)Medical Research Council (MRC) Molecular Haematology Unit, Weatherall
Institute of Molecular Medicine, Oxford University, Oxford OX3 9DS, UK.
(8)Department of Upper Gastrointestinal &Bariatric Surgery, Singapore General
Hospital, Singapore 169608, Singapore.
(9)Division of Surgical Oncology, National Cancer Centre Singapore, 11 Hospital
Drive, Singapore 169610, Singapore.
(10)Department of General Surgery, Singapore General Hospital, Singapore 169608,
Singapore.
(11)Department of Medical Oncology, Yonsei University College of Medicine, Seoul
120-752, South Korea.
(12)SingHealth/Duke-NUS Institute of Precision Medicine, National Heart Centre
Singapore, Singapore 168752, Singapore.
(13)Laboratory of Cancer Epigenome, Department of Medical Sciences, National
Cancer Centre, 11 Hospital Drive, Singapore 169610, Singapore.
(14)School of Biological Sciences, Nanyang Technological University, Singapore
637551, Singapore.
(15)Cellular and Molecular Research, National Cancer Centre, 11 Hospital Drive,
Singapore 169610, Singapore. The "hallmarks" of pancreatic ductal adenocarcinoma (PDAC) include
proliferative, invasive, and metastatic tumor cells and an associated dense
desmoplasia comprised of fibroblasts, pancreatic stellate cells, extracellular
matrix, and immune cells. The oncogenically activated pancreatic epithelium and
its associated stroma are obligatorily interdependent, with the resulting
inflammatory and immunosuppressive microenvironment contributing greatly to the
evolution and maintece of PDAC. The peculiar pancreas-specific tumor
phenotype is a consequence of oncogenes hacking the resident pancreas
regenerative program, a tissue-specific repair mechanism regulated by discrete
super enhancer networks. Defined as genomic regions containing clusters of
multiple enhancers, super enhancers play pivotal roles in cell/tissue
specification, identity, and maintece. Hence, interfering with such super
enhancer-driven repair networks should exert a disproportionately disruptive
effect on tumor versus normal pancreatic tissue. Novel drugs that directly or
indirectly inhibit processes regulating epigenetic status and integrity,
including those driven by histone deacetylases, histone methyltransferase and
hydroxylases, DNA methyltransferases, various metabolic enzymes, and bromodomain
and extraterminal motif proteins, have shown the feasibility of disrupting super
enhancer-dependent transcription in treating multiple tumor types, including
PDAC. The idea that pancreatic adenocarcinomas rely on embedded super enhancer
transcriptional mechanisms suggests a vulnerability that can be potentially
targeted as novel therapies for this intractable disease. Clin Cancer Res;
23(7); 1647-55. ©2017 AACRSee all articles in this CCR Focus section,
"Pancreatic Cancer: Challenge and Inspiration." Metabolic changes are linked to epigenetic reprogramming and play important
roles in several tumor types. PGC-1α is a transcriptional coactivator
controlling mitochondrial biogenesis and is linked to oxidative phosphorylation.
We provide evidence that melanoma models with elevated PGC-1α levels are
characteristic of the proliferative phenotype and are sensitive to bromodomain
and extra-terminal domain (BET) inhibitor treatment. A super-enhancer region
highly occupied by the BET family member BRD4 was identified for the PGC-1α
gene. BET inhibitor treatment prevented this interaction, leading to a dramatic
reduction of PGC-1α expression. Accordingly, BET inhibition diminished
respiration and mitochondrial function in cells. In vivo, melanoma models with
high PGC-1α expression strongly responded to BET inhibition by reduction of
PGC-1α and impaired tumor growth. Altogether, our findings identify epigenetic
regulatory elements that define a subset of melanomas with high sensitivity to
BET inhibition, which opens up the opportunity to define melanoma patients most
likely to respond to this treatment, depending on their tumor characteristics. Super-enhancers comprise of clusters of enhancers that are typically defined by
the ChIP-seq analysis for active histone marks. Although the biological
significance of super-enhancers is still controversial, this concept is gaining
prominence as useful characteristics of genes that play crucial roles in normal
development and pathogenesis of cancer. In various cancer cells, super-enhancers
are often associated with genes involved in carcinogenesis. For example, in
T-cell acute lymphoblastic leukemia, the oncogenic transcription factor TAL1 and
its regulatory partners (GATA3, RUNX1 and MYB) are regulated by super-enhancers;
these genes are sensitive to transcriptional inhibition, for example, via the
pharmacological approach using a small-molecule CDK7 inhibitor. This
preferential inhibition of cancer genes can also be observed for other types of
cancer. Based on these findings, we recently performed super-enhancer profiling
combined with gene expression analysis in adult T-cell leukemia/lymphoma, which
is a genetically complicated hematological maligcy, to identify critical
genes responsible for the pathogenesis. This review article aims to discuss the
concept of super-enhancers, their significance in biomedical research, and their
potential utility in elucidating the molecular pathogenesis of cancer. Super-enhancers and stretch enhancers represent classes of transcriptional
enhancers that have been shown to control the expression of cell identity genes
and carry disease- and trait-associated variants. Specifically, super-enhancers
are clusters of enhancers defined based on the binding occupancy of master
transcription factors, chromatin regulators, or chromatin marks, while stretch
enhancers are large chromatin-defined regulatory regions of at least 3,000 base
pairs. Several studies have characterized these regulatory regions in numerous
cell types and tissues to decipher their functional importance. However, the
differences and similarities between these regulatory regions have not been
fully assessed. We integrated genomic, epigenomic, and transcriptomic data from
ten human cell types to perform a comparative analysis of super and stretch
enhancers with respect to their chromatin profiles, cell type-specificity, and
ability to control gene expression. We found that stretch enhancers are more
abundant, more distal to transcription start sites, cover twice as much the
genome, and are significantly less conserved than super-enhancers. In contrast,
super-enhancers are significantly more enriched for active chromatin marks and
cohesin complex, and more transcriptionally active than stretch enhancers.
Importantly, a vast majority of super-enhancers (85%) overlap with only a small
subset of stretch enhancers (13%), which are enriched for cell type-specific
biological functions, and control cell identity genes. These results suggest
that super-enhancers are transcriptionally more active and cell type-specific
than stretch enhancers, and importantly, most of the stretch enhancers that are
distinct from super-enhancers do not show an association with cell identity
genes, are less active, and more likely to be poised enhancers. Molecular subtyping of cancer offers tremendous promise for the optimization of
a precision oncology approach to anticancer therapy. Recent advances in
pancreatic cancer research uncovered various molecular subtypes with tumors
expressing a squamous/basal-like gene expression signature displaying a worse
prognosis. Through unbiased epigenome mapping, we identified deltaNp63 as a
major driver of a gene signature in pancreatic cancer cell lines, which we
report to faithfully represent the highly aggressive pancreatic squamous subtype
observed in vivo, and display the specific epigenetic marking of genes
associated with decreased survival. Importantly, depletion of deltaNp63 in these
systems significantly decreased cell proliferation and gene expression patterns
associated with a squamous subtype and transcriptionally mimicked a subtype
switch. Using genomic localization data of deltaNp63 in pancreatic cancer cell
lines coupled with epigenome mapping data from patient-derived xenografts, we
uncovered that deltaNp63 mainly exerts its effects by activating
subtype-specific super enhancers. Furthermore, we identified a group of 45
subtype-specific super enhancers that are associated with poorer prognosis and
are highly dependent on deltaNp63. Genes associated with these enhancers
included a network of transcription factors, including HIF1A, BHLHE40, and RXRA,
which form a highly intertwined transcriptional regulatory network with
deltaNp63 to further activate downstream genes associated with poor survival. Super-enhancers (SEs) are clusters of transcriptional enhancers which control
the expression of cell identity and disease-associated genes. Current studies
demonstrated the role of multiple factors in SE formation; however, a systematic
analysis to assess the relative predictive importance of chromatin and sequence
features of SEs and their constituents is lacking. In addition, a predictive
model that integrates various types of data to predict SEs has not been
established. Here, we integrated diverse types of genomic and epigenomic
datasets to identify key signatures of SEs and investigated their predictive
importance. Through integrative modeling, we found Cdk8, Cdk9, and Smad3 as new
features of SEs, which can define known and new SEs in mouse embryonic stem
cells and pro-B cells. We compared six state-of-the-art machine learning models
to predict SEs and showed that non-parametric ensemble models performed better
as compared to parametric. We validated these models using cross-validation and
also independent datasets in four human cell-types. Taken together, our
systematic analysis and ranking of features can be used as a platform to define
and understand the biology of SEs in other cell-types. Cellular identity relies on cell-type-specific gene expression controlled at the
transcriptional level by cis-regulatory elements (CREs). CREs are unevenly
distributed across the genome, giving rise to individual CREs and clusters of
CREs (COREs). Technical and biological features hinder CORE identification. We
addressed these issues by developing an unsupervised machine learning approach
termed clustering of genomic regions analysis method (CREAM). CREAM automates
CORE detection from chromatin accessibility profiles that are enriched in CREs
strongly bound by master transcription regulators, proximal to highly expressed
and essential genes, and discriminating cell identity. Although COREs share
similarities with super-enhancers, we highlight differences in terms of the
genomic distribution and structure of these cis-regulatory units. We further
show the enhanced value of COREs over super-enhancers to identify master
transcription regulators, highly expressed and essential genes defining cell
identity. COREs enrich at topologically associated domain (TAD) boundaries. They
are also preferentially bound by the chromatin looping factors CTCF and cohesin,
in contrast to super-enhancers, forming clusters of CTCF and cohesin binding
regions and defining homotypic clusters of transcription regulator binding
regions (HCTs). Finally, we show the clinical utility of CREAM to identify COREs
across chromatin accessibility profiles to stratify more than 400 tumor samples
according to their cancer type and to delineate cancer type-specific active
biological pathways. Collectively, our results support the utility of CREAM to
delineate COREs underlying, with greater accuracy than individual CREs or
super-enhancers, the cell-type-specific biological underpinning across a wide
range of normal and cancer cell types. Super-enhancers (SE) have become a popular concept and are widely used as a
feature defining key identity genes. Here, we provide perspectives on the use of
SE to define and identify cell/tissue-identity genes. By mining SE and their
associated genes using murine functional genomics data, we highlight and discuss
current limitations and open questions regarding both the sensitivity and
specificity of identity genes/transcription factors predicted by SE. In this
context, we point to cell/tissue-specific promoters as an important additional
level of information, which we propose to combine with SE when aiming to define
potential identity genes. |
Is metoprolol metabolized by CYP2D6? | Yes, metoprolol is metabolized by CYP2D6. | Patients with cardiovascular diseases are often treated by concurrent multiple
drug therapy. It is therefore plausible that with an increasing number of drugs
the risk of drug interactions increases. Such interactions can be either
pharmacodynamic (and are due to the mechanism of the administered drugs) or they
can be pharmacokinetic (resulting in a reduction or enhancement of drug
elimination). Pharmacokinetic interactions can be either due to interactions at
the level of drug metabolizing enzymes (most important cytochrome P450 (CYP)
enzymes) or interactions at the level of drug transporter proteins (for example
P-glycoprotein (MDR1)). It is important to distinguish between both mechanisms
because interactions at transporter proteins can be attributed to those drugs
that are not enzymatically metabolized. The scope of this article is to give an
overview on clinically relevant interactions of the four beta-blockers widely
used in the therapy of cardiovascular diseases namely atenolol (CAS 29122-68-7),
bisoprolol (CAS 66722-44-9), metoprolol (CAS 37350-58-6) (each beta-1
selective), and carvedilol (CAS 72956-09-3) (beta-1 and beta-2 nonselective).
Among these beta-blockers atenolol is mainly eliminated by renal excretion,
bisoprolol is in part excreted as parent compound via the renal route (50%), the
other 50% are hepatically metabolised, whereas metoprolol and carvedilol are
metabolised by CYP2D6. In addition, evidence is accumulating that carvedilol is
a substrate for P-glycoprotein. For these four beta-blockers various
pharmacodynamic and pharmacokinetic interactions have been demonstrated. Such
interactions that result in an altered pharmacokinetics are mainly observed with
those beta-blockers that are excreted via metabolism (metoprolol and
carvedilol). Accordingly these drugs have a higher potential for drug
interactions. However, it should be emphasized that, in general, beta-blockers
are well tolerated safe drugs with a large therapeutic index. |
List versions of ExpansionHunter | ExpansionHunter and ExpansionHunter Denovo | SUMMARY: We describe a novel computational method for genotyping repeats using
sequence graphs. This method addresses the long-standing need to accurately
genotype medically important loci containing repeats adjacent to other variants
or imperfect DNA repeats such as polyalanine repeats. Here we introduce a new
version of our repeat genotyping software, ExpansionHunter, that uses this
method to perform targeted genotyping of a broad class of such loci.
AVAILABILITY AND IMPLEMENTATION: ExpansionHunter is implemented in C++ and is
available under the Apache License Version 2.0. The source code, documentation,
and Linux/macOS binaries are available at
https://github.com/Illumina/ExpansionHunter/.
SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics
online. |
What are the uses of Nirsevimab? | A single injection of nirsevimab resulted in fewer medically attended RSV-associated lower respiratory tract infections and hospitalizations than placebo throughout the RSV season in healthy preterm infants. | BACKGROUND: Respiratory syncytial virus (RSV) is the most common cause of lower
respiratory tract infection in infants, and a need exists for prevention of RSV
in healthy infants. Nirsevimab is a monoclonal antibody with an extended
half-life that is being developed to protect infants for an entire RSV season
with a single intramuscular dose.
METHODS: In this trial conducted in both northern and southern hemispheres, we
evaluated nirsevimab for the prevention of RSV-associated lower respiratory
tract infection in healthy infants who had been born preterm (29 weeks 0 days to
34 weeks 6 days of gestation). We randomly assigned the infants in a 2:1 ratio
to receive nirsevimab, at a dose of 50 mg in a single intramuscular injection,
or placebo at the start of an RSV season. The primary end point was medically
attended RSV-associated lower respiratory tract infection through 150 days after
administration of the dose. The secondary efficacy end point was hospitalization
for RSV-associated lower respiratory tract infection through 150 days after
administration of the dose.
RESULTS: From November 2016 through November 2017, a total of 1453 infants were
randomly assigned to receive nirsevimab (969 infants) or placebo (484 infants)
at the start of the RSV season. The incidence of medically attended
RSV-associated lower respiratory tract infection was 70.1% lower (95% confidence
interval [CI], 52.3 to 81.2) with nirsevimab prophylaxis than with placebo (2.6%
[25 infants] vs. 9.5% [46 infants]; P<0.001) and the incidence of
hospitalization for RSV-associated lower respiratory tract infection was 78.4%
lower (95% CI, 51.9 to 90.3) with nirsevimab than with placebo (0.8% [8 infants]
vs. 4.1% [20 infants]; P<0.001). These differences were consistent throughout
the 150-day period after the dose was administered and across geographic
locations and RSV subtypes. Adverse events were similar in the two trial groups,
with no notable hypersensitivity reactions.
CONCLUSIONS: A single injection of nirsevimab resulted in fewer medically
attended RSV-associated lower respiratory tract infections and hospitalizations
than placebo throughout the RSV season in healthy preterm infants. (Funded by
AstraZeneca and Sanofi Pasteur; ClinicalTrials.gov number, NCT02878330.). |
What is the proteoglycan Tsukushi? | Tsukushi (TSK), a member of the small leucine-rich repeat proteoglycan (SLRP) family, plays multifunctional roles by interacting with signaling molecules during development.
In lung cancer cells, TSK is expressed more highly than the other SLRPs family members, and regulates the EMT and proliferation. Thus, TSK may be a key coordinator of multiple pathways and an important structural element in the lung cancer microenvironment.
Gain- and loss-of-function analyses showed that the small leucine-rich proteoglycan, tsukushi, contributes to vitamin K2-mediated enhancement of collagen accumulation. | Vitamin K2 is a critical nutrient required for blood coagulation. It also plays
a key role in bone homeostasis and is a clinically effective therapeutic agent
for osteoporosis. We previously demonstrated that vitamin K2 is a
transcriptional regulator of bone marker genes in osteoblastic cells and that it
may potentiate bone formation by activating the steroid and xenobiotic receptor,
SXR. To explore the SXR-mediated vitamin K2 signaling network in bone
homeostasis, we identified genes up-regulated by both vitamin K2 and the
prototypical SXR ligand, rifampicin, in osteoblastic cells using oligonucleotide
microarray analysis and quantitative reverse transcription-PCR. Fourteen genes
were up-regulated by both ligands. Among these, tsukushi, matrilin-2, and CD14
antigen were shown to be primary SXR target genes. Moreover, collagen
accumulation in osteoblastic MG63 cells was enhanced by vitamin K2 treatment.
Gain- and loss-of-function analyses showed that the small leucine-rich
proteoglycan, tsukushi, contributes to vitamin K2-mediated enhancement of
collagen accumulation. Our results suggest a new function for vitamin K2 in bone
formation as a transcriptional regulator of extracellular matrix-related genes,
that are involved in the collagen assembly. The nuclear receptor steroid and xenobiotic receptor (SXR) is a transcriptional
regulator activated by various biological and xenobiotic substances. We have
recently shown that SXR is expressed in bone and that this receptor is critical
for bone metabolism, particularly in osteoblastic cells. Vitamin K2, one of the
critical nutrients in bone metabolism, has been demonstrated that it is a potent
SXR agonist and modulates the expression of various bone-related genes in
osteoblastic cells. Using microarray analysis, we identified novel SXR target
genes that were activated by vitamin K2 in osteoblastic cells. Among them, a
small leucine-rich repeat proteoglycan, tsukushi, has been shown to contribute
to collagen accumulation, and the protein may interact with another vitamin
K2-inducible SXR target, matrilin-2, a member of the matrilin family that
functions as collagen adaptors. Besides functioning as a xenobiotic biosensor,
our findings show that SXR is also a vitamin K2 target and an important
transcriptional factor that regulates bone homeostasis in bone cells. |
Does Curare function by stimulating the acetylcholine receptor? | No. Curare function does not stimulate the acetylcholine receptor. | We have studied the effects of curare on responses resulting from iontophoretic
application of several putative neurotransmitters onto Aplysia neurons. These
neurons have specific receptors for acetylcholine (ACh), dopamine, octopamine,
phenylethanolamine, histamine, gamma-aminobutyric acid (GABA), aspartic acid,
and glutamic acid. Each of these substances may on different specific neurons
elicit at least three types of response, caused by a fast depolarizing Na+, a
fast hyperpolarizing Cl-, or a slow hyperpolarizing K+ conductance increase. All
responses resulting from either Na+ or Cl- conductance increases, irrespective
of which putative transmitter activated the response, were sensitive to curare.
Most were totally blocked by less than or equal to 10-4 M curare. GABA responses
were less sensitive and were often only depressed by 10-3 M curare. K+
conductance responses, irrespective of the transmitter, were not curare
sensitive. These results are consistent with a model of receptor organization in
which one neurotransmitter receptor may be associated with any of at least three
ionophores, mediating conductance increase responses to Na+, Cl-, and K+,
respectively. In Aplysia nervous tissue, curare appears not to be a specific
antagonist for the nicotinic ACh receptor, but rather to be a specific blocking
agent for a class of receptor-activated Na+ and Cl- responses. The perhydro derivative of histrionicotoxin reversibly blocks the excitatory
ionic transduction system in the synaptic and sarcolemmal membranes of mammalian
skeletal muscle cells. The efficacy of perhydrohistrionicotoxin as an antagonist
at the post-synaptic membrane is increased by the transient presence of
acetylcholine in the endplate of innervated muscles and at extrajunctional
receptors in denervated muscles. alpha-Bungarotoxin and
[(3)H]monoacetyl-alpha-bungarotoxin block the endplate acetylcholine receptors,
each binding to the same extent. The effect of bungarotoxin is partially
reversible. These electrophysiological results, together with the effects of
perhydrohistrionicotoxin and/or d-tubocurarine on the binding of
[(3)H]monoacetyl-alpha-bungarotoxin at endplates of murine diaphragm muscle and
on the bungarotoxin-elicited irreversible blockade of neuromuscular
transmission, suggest that at least two types of sites participate in the
synaptic excitation by acetylcholine. One site, competitively blocked by
bungarotoxin and by curare, is presumably the acetylcholine receptor. Binding of
bungarotoxin at this site is responsible for an irreversible blockade of
neuromuscular transmission. The second site, competitively blocked by
bungarotoxin and perhydrohistrionicotoxin, is proposed to be part of the
cholinergic ion conductance modulator. Binding of bungarotoxin to this site does
not result in an irreversible blockade. The mode of action of curare, a well-known competitive antagonist of
acetylcholine at the nicotinic receptor, was examined with the single channel
recording technique. Curare can open cholinergic channels in rat myotubes, as
suggested by Ziskind and Dennis (1978). Moreover another curare molecule can
then block the curare-activated open channel, in line with previous results
concerning such a mode of action. In adult rat muscle, the partial agonist
activity of curare can also be demonstrated, though it is much weaker than in
embryonic muscle. It is also shown that in adult muscle cell, the conductance of
the channel (activated either by ACh or by curare) is 50-60 pS, i.e., higher
than in the myotubes (35 pS). d-Tubocurarine (curare) is a well-characterized competitive antagonist of
nicotinic acetylcholine receptors (AChRs), and it is usually assumed that curare
and agonists share a common binding site. We have examined the role of several
highly conserved residues of the alpha-, gamma-, and delta-subunits in the
interaction of curare with the Torpedo acetylcholine receptor (AChR). Curare
inhibition of wild-type receptors is consistent with curare binding to a single
high-affinity binding site [inhibitor constant (Ki) = 20 nM]. Phenylalanine
substitutions for two tyrosine residues implicated as being in the ligand
binding site (alpha Y93F, alpha Y190F) reduce curare affinity, indicating that
these residues are also important for high-affinity curare binding.
Phenylalanine substitution for alpha Y198 [alpha Y198F (notation used here:
subunit/amino acid in wild-type/residue number/substitution)] causes a 10-fold
increase in curare affinity (Ki = 3.1 nM), and measurement of the recovery from
curare inhibition indicates that this increase in affinity is due to a reduction
in the rate of curare dissociation from the receptor. In addition to the
alpha-subunits, portions of the ligand binding sites also reside on the gamma-
and delta-subunits, and photoaffinity studies have implicated two residues
(gamma W55 and delta W57) as forming part of the curare sites. The gamma W55L
mutation results in an eightfold decrease in curare affinity (Ki = 170 nM),
whereas the delta W57L mutation has no effect. These data support the notion
that the high-affinity curare binding site is formed by segments of the alpha-
and gamma-subunits.(ABSTRACT TRUNCATED AT 250 WORDS) Spinal motoneurons (MNs) in the chick embryo undergo programmed cell death
coincident with the establishment of nerve-muscle connections and the onset of
synaptic transmission at the neuromuscular junction. Chronic treatment of
embryos during this period with nicotinic acetylcholine receptor
(nAChR)-blocking agents [e.g., curare or alpha-bungarotoxin (alpha-BTX)]
prevents the death of MNs. Although this rescue effect has been attributed
previously to a peripheral site of action of the nAChR-blocking agents at the
neuromuscular junction (NMJ), because nAChRs are expressed in both muscle and
spinal cord, it has been suggested that the rescue effect may, in fact, be
mediated by a direct central action of nAChR antagonists. By using a variety of
different nAChR-blocking agents that target specific muscle or neuronal nAChR
subunits, we find that only those agents that act on muscle-type receptors block
neuromuscular activity and rescue MNs. However, paralytic, muscular dysgenic
mutant chick embryos also exhibit significant increases in MN survival that can
be further enhanced by treatment with curare or alpha-BTX, suggesting that
muscle paralysis may not be the sole factor involved in MN survival. Taken
together, the data presented here support the argument that, in vivo, nAChR
antagonists promote the survival of spinal MNs primarily by acting peripherally
at the NMJ to inhibit synaptic transmission and reduce or block muscle activity.
Although a central action of these agents involving direct perturbations of MN
activity may also play a contributory role, further studies are needed to
determine more precisely the relative roles of central versus peripheral sites
of action in MN rescue. Nicotinic acetylcholine receptors are members of the ligand-gated ion channel
superfamily, that includes also gamma-amino-butiric-acid(A), glycine, and
5-hydroxytryptamine(3) receptors. Functional nicotinic acetylcholine receptors
result from the association of five subunits each contributing to the pore
lining. The major neuronal nicotinic acetylcholine receptors are heterologous
pentamers of alpha4beta2 subunits (brain), or alpha3beta4 subunits (autonomic
ganglia). Another class of neuronal receptors that are found both in the central
and peripheral nervous system is the homomeric alpha7 receptor. The muscle
receptor subtypes comprise of alphabetadeltagamma (embryonal) or
alphabetadeltaepsilon (adult) subunits. Although nicotinic acetylcholine
receptors are not directly involved in the hypnotic component of anesthesia, it
is possible that modulation of central nicotinic transmission by volatile agents
contributes to analgesia. The main effect of anesthetic agents on nicotinic
acetylcholine receptors is inhibitory. Volatile anesthetics and ketamine are the
most potent inhibitors both at alpha4beta2 and alpha3beta4 receptors with
clinically relevant IC(50) values. Neuronal nicotinic acetylcholine receptors
are more sensitive to anesthetics than their muscle counterparts, with the
exception of the alpha7 receptor. Several intravenous anesthetics such as
barbiturates, etomidate, and propofol exert also an inhibitory effect on the
nicotinic acetylcholine receptors, but only at concentrations higher than those
necessary for anesthesia. Usual clinical concentrations of curare cause
competitive inhibition of muscle nicotinic acetylcholine receptors while higher
concentrations may induce open channel blockade. Neuronal nAChRs like
alpha4beta2 and alpha3beta4 are inhibited by atracurium, a curare derivative,
but at low concentrations the alpha4beta2 receptor is activated. Inhibition of
sympathetic transmission by clinically relevant concentrations of some
anesthetic agents is probably one of the factors involved in arterial
hypotension during anesthesia. To elucidate innervation in the upper esophageal sphincter (UES) muscle of the
eel, a key muscle in swallowing, repetitive electrical field stimulation (EFS;
30 mA, 40 V, 300 micros, 10 Hz, 10 trains) was employed. Anatomically, the eel
UES muscle consists of striated fibers. The EFS-induced contraction of the UES
was completely blocked by tetrodotoxin and curare, and abolished in Ca2+ -free
Ringer solution. These results suggest that the EFS stimulates nerve fibers
specifically and releases acetylcholine as a neurotransmitter. In fact,
acetylcholine and carbachol constricted the UES in a concentration-dependent
manner. Even after blocking neuronal firing with tetrodotoxin, acetylcholine
constricted the UES muscle, suggesting the existence of acetylcholine receptors
on the UES muscle cells. Both EFS- and carbachol-evoked contractions of the UES
were blocked by curare at a lower concentration than by atropine or
hexamethonium, suggesting that the acetylcholine receptor is nicotinic. Even in
Ca2+ -free Ringer solution, a direct current stimulus (2 s duration) constricted
the UES muscle to an extent similar to that in the presence of Ca2+, indicating
that the muscle contraction itself does not need extracellular Ca2+, i.e., the
muscle can be constricted by a release of Ca2+ from the sarcoplasmic reticulum. Erabutoxins a and b are neurotoxins isolated from venom of a sea snake Laticauda
semifasciata (erabu-umihebi). Amino acid sequences of the toxins indicated that
the toxins are members of a superfamily consisting of short and long neurotoxins
and cytotoxins found in sea snakes and terrestrial snakes. The short neurotoxins
to which erabutoxins belong act by blocking the nicotinic acetylcholine receptor
on the post synaptic membrane in a manner similar to that of curare. X-ray
crystallography and NMR analyses showed that the toxins have a three-finger
structure, in which three fingers made of three loops emerging from a dense core
make a gently concave surface of the protein. The sequence comparison and the
location of essential residues on the protein suggested the mechanism of binding
of the toxin to the acetylcholine receptor. Classification of snakes by means of
sequence comparison and that based on different morphological features were
inconsistent, which led the authors to propose a hypothesis "Evolution without
divergence." The synaptic vesicle is the essential organelle of the synapse. Many approaches
for studying synaptic vesicle recycling have been devised, one of which, the
styryl (FM) dye, is well suited for this purpose. FM dyes reversibly stain, but
do not permeate, membranes; hence they can specifically label membrane-bound
organelles. Their quantum yield is drastically higher when bound to membranes
than when in aqueous solution. This protocol describes the imaging of synaptic
vesicle recycling by staining and destaining vesicles with FM dyes. Nerve
terminals are stimulated (electrically or by depolarization with high K(+)) in
the presence of dye, their vesicles are then allowed to recycle, and finally dye
is washed from the chamber. In neuromuscular junction (NMJ) preparations,
movements of the muscle must be inhibited if imaging during stimulation is
desired (e.g., by application of curare, a potent acetylcholine receptor
inhibitor). The main characteristics of FM dyes are also reviewed here, as are
recent FM dye monitoring techniques that have been used to investigate the
kinetics of synaptic vesicle fusion. Prenatal nicotine exposure with continued exposure through breast milk over the
first week of life (developmental nicotine exposure, DNE) alters the development
of brainstem circuits that control breathing. Here, we test the hypothesis that
DNE alters the respiratory motor response to endogenous and exogenous
acetylcholine (ACh) in neonatal rats. We used the brainstem-spinal cord
preparation in the split-bath configuration, and applied drugs to the brainstem
compartment while measuring the burst frequency and amplitude of the fourth
cervical ventral nerve roots (C4VR), which contain the axons of phrenic
motoneurons. We applied ACh alone; the nicotinic acetylcholine receptor (nAChR)
antagonist curare, either alone or in the presence of ACh; and the muscarinic
acetylcholine receptor (mAChR) antagonist atropine, either alone or in the
presence of ACh. The main findings include: (1) atropine reduced frequency
similarly in controls and DNE animals, while curare caused modest slowing in
controls but no consistent change in DNE animals; (2) DNE greatly attenuated the
increase in C4VR frequency mediated by exogenous ACh; (3) stimulation of nAChRs
with ACh in the presence of atropine increased frequency markedly in controls,
but not DNE animals; (4) stimulation of mAChRs with ACh in the presence of
curare caused a modest increase in frequency, with no treatment group
differences. DNE blunts the response of the respiratory central pattern
generator to exogenous ACh, consistent with reduced availability of functionally
competent nAChRs; DNE did not alter the muscarinic control of respiratory motor
output. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1138-1149, 2016. |
Are somatic mutations positioned towards the nuclear periphery? | lamina-associated regions, which are typically localized at the nuclear periphery, displayed higher somatic mutation frequencies than did the interlamina regions at the nuclear core. Smoking and UV-related signatures, as well as substitutions at certain motifs, were more enriched in the nuclear periphery. | The nuclear matrix (NM) is the structural framework of the nucleus that consists
of the peripheral lamins and pore complexes, an internal ribonucleic protein
network, and residual nucleoli. Differences between the nuclear matrix protein
(NMP) composition of transformed cells and their normal homologues were detected
in numerous cases. Actually several tumor-specific nuclear matrix proteins
(NMPs) are proposed for diagnostic of bladder, breast, colon and some other
cancers. According to the role of NMPs in development and phenotype of a given
neoplasms the tumors can be classified as follows: I. Tumors bearing mutations
in the genes encoding NMPs. The group consists of following subgroups: 1)
hereditary cancer syndromes with mutations in the NM-attached oncoproteins or
tumor suppressor genes; 2) sporadic tumors with somatic mutations in the
NM-attached oncoproteins, tumor suppressor genes or replication enzymes; 3)
leukemias with fused NMPs. II. Tumors with phenotypic quantitative or
qualitative changes of the NMP spectrum. Nuclear organization of genomic DNA affects processes of DNA damage and repair,
yet its effects on mutational landscapes in cancer genomes remain unclear. Here
we analyzed genome-wide somatic mutations from 366 samples of six cancer types.
We found that lamina-associated regions, which are typically localized at the
nuclear periphery, displayed higher somatic mutation frequencies than did the
interlamina regions at the nuclear core. This effect was observed even after
adjustment for features such as GC percentage, chromatin, and replication
timing. Furthermore, mutational signatures differed between the nuclear core and
periphery, thus indicating differences in the patterns of DNA-damage or
DNA-repair processes. For instance, smoking and UV-related signatures, as well
as substitutions at certain motifs, were more enriched in the nuclear periphery.
Thus, the nuclear architecture may influence mutational landscapes in cancer
genomes beyond the previously described effects of chromatin structure and
replication timing. |
Which biological drugs are EMA approved for pediatric psoriasis? | Currently there are three European Medicines Agency (EMA)-approved biological treatment options for pediatric psoriasis: etanercept, ustekinumab, and adalimumab. | Background and Objectives: Severe, recalcitrant cases of pediatric psoriasis or
atopic dermatitis may necessitate treatment with biological agents; however,
this may be difficult due to lack of treatment options and standardized
treatment guidelines. This review evaluates the biological treatment options
available, including off-label uses, and provides a basic therapeutic guideline
for pediatric psoriasis and atopic dermatitis. Materials and Methods: A PubMed
review of biological treatments for pediatric psoriasis and atopic dermatitis
with information regarding age, efficacy, dosing, contra-indications, adverse
events, and off-label treatments. Results: Currently there are three European
Medicines Agency (EMA)-approved biological treatment options for pediatric
psoriasis: etanercept, ustekinumab, and adalimumab. While dupilumab was recently
Food and Drug Administration (FDA)- and EMA-approved for adult atopic
dermatitis, it is still not yet approved for pediatric atopic dermatitis.
Conclusions: Given the high morbidity associated with pediatric atopic
dermatitis and psoriasis, there is a need for more treatment options. Further
research and post-marketing registries are needed to extend the use of biologics
into pediatric patients. |
What is the role of Adamts18 in hormone receptor signaling? | Adamts18 links luminal hormone receptor signaling to basement membrane remodeling and stem cell activation. | |
Is belimumab effective for the lupus nephritis? | Yes, belimumab appears to effective for the lupus nephritis. | We report the case of a 19-year-old woman with progressive proliferative lupus
nephritis (LN) class III after induction and maintece therapy with
mycophenolate mofetil (MMF). Despite a satisfying clinical improvement
proteinuria progressed under this medication. We treated the patient with
additional belimumab after discussing other options. Following treatment with
belimumab, proteinuria rapidly improved to almost normal levels and clinical
remission lasted. Belimumab might hold promise for this indication. Recently introduced into the market, belimumab (Benlysta) is a monoclonal
antibody that has potential clinically efficacious applications for the
treatment of lupus nephritis. Lupus nephritis is a major complication of
systemic lupus erythematosus (SLE) that can lead to significant illness or even
death without proper intervention and treatment. With vast implications through
a novel mechanism, belimumab offers a new standard of treatment for physicians
in the complications associated with SLE, specifically lupus nephritis. By
targeting B cell signaling and maturation, belimumab is able to mitigate the
underlying pathological complications surrounding SLE. Phase 3 clinical trials
with belimumab have depicted clinically efficacious applications, suggesting
belimumab as a revolutionary breakthrough in the treatment armamentarium for
practicing clinicians. This article explains the precise mechanism of action of
belimumab on the soluble protein BlyS that plays a major role in the
pathogenesis of lupus nephritis. In addition, the extensive pharmacokinetics and
clinical implications are exemplified in this review with belimumab's comparison
with standard therapeutic guidelines for the treatment of lupus nephritis. BACKGROUND: The treatment of Lupus Nephritis (LN) is an unmet need in the
management of patients with Systemic Lupus Erythematosus (SLE).
CASE PRESENTATION : We report two cases of women affected by Lupus Nephritis
(LN) ISN/RNP Class IV with serological active disease, high disease activity and
marked fatigue. In both cases, Mycophenolate mofetil (MMF), as induction
therapy, was poorly tolerated because of gastrointestinal toxicity. Belimumab,
together with low-doses of MMF, was effective as induction treatment leading to
early achievement of complete renal response in these two selected cases of LN.
CONCLUSIONS: We also report a literature review concerning the efficacy and
safety of Belimumab in Lupus Nephritis. Further studies are needed to evaluate
the use of Belimumab to manage the renal involvement in patients with Systemic
Lupus Erythematosus, waiting for the results of ongoing randomized clinical
trials. Background and Objectives: Belimumab (BEL) is a monoclonal antibody approved for
the treatment of active systemic lupus erythematosus (SLE) but not for lupus
nephritis (LN) and neuropsychiatric systemic lupus erythematosus (NPSLE). We
aimed to assess BEL's effects on these severe, potentially life-threatening
manifestations. Methods: Retrospective observational cohort study using routine
clinical data in a case series of patients with SLE receiving BEL. Results:
Sixteen patients received BEL therapy for active SLE. Nine were excluded because
they had no LN or NPSLE. Six suffered from LN, and one patient had NPSLE. All LN
patients received BEL in addition to standard therapy including glucocorticoids,
hydroxychloroquine, and mycophenolate mofetil in five cases, and tacrolimus in
one case. Three patients with proteinuria >1,000 mg/g creatinine responded well
(one complete, two partial renal responses); all other patients had decreasing
proteinuria and a reduction in anti-dsDNA levels. The patient with NPSLE who had
failed previous therapies had persistent clinical improvement of cutaneous and
neuropsychiatric manifestations. There was one mild allergic reaction and one
lower respiratory tract infection, but no other adverse events. One patient
discontinued therapy due to a lack of improvement in clinical symptoms, another
because of clinical remission. Conclusions: In our series, BEL led to a decrease
of proteinuria in patients with proteinuria of more than 1,000 mg/g creatinine
despite standard of care treatment, and led to a marked clinical improvement in
one patient with NPSLE. No adverse events were observed. Routinely administered
BEL shows clinical efficacy on non-approved manifestations, but careful patient
selection is warranted. BACKGROUND: Anti-CD20 B-cell depletion has not shown superior efficacy to
standard immunosuppression in patients with systemic lupus erythematosus (SLE).
Besides trial design, potential explanations are incomplete B-cell depletion in
relation to substantial surges in B-cell-activating factor (BAFF). To improve
B-cell targeting strategies, we conducted the first study in SLE patients aimed
at investigating immunological effects and feasibility of combining rituximab
(RTX; anti-CD20) and belimumab (BLM; anti-BAFF).
METHODS: Reported is the long-term follow-up of a Phase 2 proof-of-concept study
in 15 patients with SLE including 12 (80%) with lupus nephritis (LN).
RESULTS: In 10/15 (67%) patients, a clinical response was observed by
achievement of lupus low disease activity state, of which 8 (53%) continued
treatment (BLM + ≤7.5 mg prednisolone) for the complete 2 years of follow-up.
Five patients (33%) were referred to as 'non-responders' due to persistent LN,
major flare or repetitive minor flares. Out of 12 LN patients, 9 (75%) showed a
renal response including 8 (67%) complete renal responders. All anti-dsDNA+
patients converted to negative, and both anti-C1q and extractable nuclear
antigen autoantibodies showed significant reductions. CD19+ B cells showed a
median decrease from baseline of 97% at 24 weeks, with a persistent reduction of
84% up to 104 weeks. When comparing responders with non-responders, CD20+ B
cells were depleted significantly less in non-responders and double-negative
(DN) B cells repopulated significantly earlier.
CONCLUSIONS: Combined B-cell targeted therapy with RTX and BLM prevented full
B-cell repopulation including DN B cells, with concomitant specific reduction of
SLE-relevant autoantibodies. The observed immunological and clinical benefits in
a therapy-refractory SLE population prompt further studies on RTX + BLM. BACKGROUND: In adults with active lupus nephritis, the efficacy and safety of
intravenous belimumab as compared with placebo, when added to standard therapy
(mycophenolate mofetil or cyclophosphamide-azathioprine), are unknown.
METHODS: In a phase 3, multinational, multicenter, randomized, double-blind,
placebo-controlled, 104-week trial conducted at 107 sites in 21 countries, we
assigned adults with biopsy-proven, active lupus nephritis in a 1:1 ratio to
receive intravenous belimumab (at a dose of 10 mg per kilogram of body weight)
or matching placebo, in addition to standard therapy. The primary end point at
week 104 was a primary efficacy renal response (a ratio of urinary protein to
creatinine of ≤0.7, an estimated glomerular filtration rate [eGFR] that was no
worse than 20% below the value before the renal flare (pre-flare value) or ≥60
ml per minute per 1.73 m2 of body-surface area, and no use of rescue therapy),
and the major secondary end point was a complete renal response (a ratio of
urinary protein to creatinine of <0.5, an eGFR that was no worse than 10% below
the pre-flare value or ≥90 ml per minute per 1.73 m2, and no use of rescue
therapy). The time to a renal-related event or death was assessed.
RESULTS: A total of 448 patients underwent randomization (224 to the belimumab
group and 224 to the placebo group). At week 104, significantly more patients in
the belimumab group than in the placebo group had a primary efficacy renal
response (43% vs. 32%; odds ratio, 1.6; 95% confidence interval [CI], 1.0 to
2.3; P = 0.03) and a complete renal response (30% vs. 20%; odds ratio, 1.7; 95%
CI, 1.1 to 2.7; P = 0.02). The risk of a renal-related event or death was lower
among patients who received belimumab than among those who received placebo
(hazard ratio, 0.51; 95% CI, 0.34 to 0.77; P = 0.001). The safety profile of
belimumab was consistent with that in previous trials.
CONCLUSIONS: In this trial involving patients with active lupus nephritis, more
patients who received belimumab plus standard therapy had a primary efficacy
renal response than those who received standard therapy alone. (Funded by
GlaxoSmithKline; BLISS-LN ClinicalTrials.gov number, NCT01639339.). PURPOSE OF REVIEW: Despite ground-breaking innovations for most autoimmune
diseases, the treatment of lupus nephritis has remained largely the same for
decades because none of the tested drugs demonstrated superiority over
standard-of-care in randomized controlled clinical trials.
RECENT FINDINGS: Recently, the Belimumab in Subjects with Systemic Lupus
Erythematosus - Lupus Nephritis trial tested belimumab, an inhibitor of B-cell
activating factor, as an add-on therapy to steroids and either mycophenolate
mofetil (MMF) or cyclophosphamide when given IV monthly over a period of 104
weeks at an effect size of 11% for a Primary Efficacy Renal Response. The
NOBILITY trial reported positive results for the B-cell-depleting agent
obinutuzumab as an add-on therapy to steroids and MMF when given IV every 6
months over a period of 76 weeks at an effect size of 22% for a complete renal
response (CRR). The AURORA trial reported positive results for the calcineurin
inhibitor voclosporin as an oral add-on therapy to low dose steroids and MMF
when given twice daily over a period of 52 weeks at an effect size of 18.5% for
a CRR.
SUMMARY: These studies will change the treatment landscape of lupus nephritis.
In which way is discussed in this article. |
Which are the lactate isomers? | Lactate contains a chiral carbon and thus has two optical isomers-d-lactate and l-lactate. | Lactate contains a chiral carbon and thus has two optical isomers-d-lactate and
l-lactate. l-Lactate is the predomit form that is produced by the body and
can be delivered to the organs. On the other hand, gut microbiota produce both
isomers, which can then flow into the body. Although both d-lactate and
l-lactate can contribute to energy metabolism, their potential roles in
adipocyte differentiation remain to be elucidated. Here, we investigated the
effects of l-lactate and d-lactate on the differentiation of 3T3-L1
preadipocytes. Both lactate etiomers were demonstrated to enhance
triglyceride accumulation by stimulating the early phase of adipocyte
differentiation. Notably, d-lactate was more potent than l-lactate in inducing
triglyceride accumulation. The degree of triglyceride accumulation induced by
l-lactate was similar to that induced by pyruvate. d-Lactate was more potent
than l-lactate in increasing the activity of glycerol-3-phosphate dehydrogenase.
Both lactate etiomers did not affect cell viability. Moreover, both
etiomers upregulated the expression of peroxisome proliferator-activated
receptor γ, CCAAT/enhancer-binding protein (C/EBP) α, sterol regulatory
element-binding protein-1c, and fatty acid synthase, with d-lactate exerting
stronger effects than l-lactate. By contrast, lactate did not influence the
expression of C/EBPβ and C/EBPδ. d-Lactate significantly increased and l-lactate
tended to increase p38 MAPK phosphorylation, and the p38 MAPK inhibitor SB203580
inhibited the stimulation of adipocyte differentiation by d-lactate and
l-lactate. These findings showed that both lactate etiomers stimulate
preadipocyte differentiation, with d-lactate showing more potent effects than
l-lactate. In addition, our study demonstrated that d-lactate and l-lactate
exert different effects on physiological events. |
What is the function of the Eyeless associated gene in Drosophila? | Eyeless (ey) also known as Pax6, is one of the most critical transcription factors for initiating the entire eye development in Drosophila. | The Drosophila gene eyeless (ey) encodes a transcription factor with both a
paired domain and a homeodomain. It is homologous to the mouse Small eye (Pax-6)
gene and to the Aniridia gene in humans. These genes share extensive sequence
identity, the position of three intron splice sites is conserved, and these
genes are expressed similarly in the developing nervous system and in the eye
during morphogenesis. Loss-of-function mutations in both the insect and in the
mammalian genes have been shown to lead to a reduction or absence of eye
structures, which suggests that ey functions in eye morphogenesis. By targeted
expression of the ey complementary DNA in various imaginal disc primordia of
Drosophila, ectopic eye structures were induced on the wings, the legs, and on
the antennae. The ectopic eyes appeared morphologically normal and consisted of
groups of fully differentiated ommatidia with a complete set of photoreceptor
cells. These results support the proposition that ey is the master control gene
for eye morphogenesis. Because homologous genes are present in vertebrates,
ascidians, insects, cephalopods, and nemerteans, ey may function as a master
control gene throughout the metazoa. Development of the vertebrate eye requires a series of steps including
specification of the anterior neural plate, evagination of the optic vesicles
from the ventral forebrain, and the cellular differentiation of the lens and
retina. Homeobox-containing genes, especially the transcription regulator Pax6,
play a critical role in vertebrate and invertebrate eye formation. Mutations in
Pax6 function result in eye malformations known as Aniridia in humans and Small
eye syndrome in mice. The Drosophila homologue of Pax6, eyeless, is also
necessary for correct invertebrate eye development, and its misexpression leads
to formation of ectopic eyes in Drosophila. Here we show that a conserved
vertebrate homeobox gene, Rx, is essential for normal eye development, and that
its misexpression has profound effects on eye morphology. Xenopus embryos
injected with synthetic Rx RNA develop ectopic retinal tissue and display
hyperproliferation in the neuroretina. Mouse embryos carrying a null allele of
this gene do not form optic cups and so do not develop eyes. The Rx gene family
plays an important role in the establishment and/or proliferation of retinal
progenitor cells. The development of the Drosophila compound eye requires the function of a set of
evolutionarily conserved genes. Among these, the Drosophila Pax-6 gene eyeless
(ey) plays a major role. ey has been considered a master control gene of eye
development in the animal kingdom because targeted expression of ey and
vertebrate as well as invertebrate homologs lead to the formation of ectopic
eyes in Drosophila. We demonstrate that an intron of the ey gene contains an
enhancer that regulates the eye specific expression of the gene in the eye disc
primordia of embryos and in the eye imaginal discs of third instar larvae.
Moreover, a 212-bp enhancer element is necessary and sufficient for the enhancer
function. It is partially conserved in Drosophila hydei and contains putative
Pax-6 Paired domain binding sites. We show that several binding sites are
required for the eye specific expression, and, therefore, we propose a
Pax-6-like molecule to be a positive transactivator for the eye specific ey
expression. This transactivator recently has been identified as twin of eyeless,
the second Pax-6 gene in Drosophila. We describe here the role of the transcription factors encoding genes tailless
(tll), atonal (ato), sine oculis (so), eyeless (ey) and eyes absent (eya), and
EGFR signaling in establishing the Drosophila embryonic visual system. The
embryonic visual system consists of the optic lobe primordium, which, during
later larval life, develops into the prominent optic lobe neuropiles, and the
larval photoreceptor (Bolwig's organ). Both structures derive from a
neurectodermal placode in the embryonic head. Expression of tll is normally
confined to the optic lobe primordium, whereas ato appears in a subset of
Bolwig's organ cells that we call Bolwig's organ founders. Phenotypic analysis,
using specific markers for Bolwig's organ and the optic lobe, of tll loss- and
gain-of-function mutant embryos reveals that tll functions to drive cells to
optic lobe as opposed to Bolwig's organ fate. Similar experiments indicate that
ato has the opposite effect, namely driving cells to a Bolwig's organ fate.
Since we can show that tll and ato do not regulate each other, we propose a
model wherein tll expression restricts the ability of cells to respond to
signaling arising from ato-expressing Bolwig's organ pioneers. Our data further
suggest that the Bolwig's organ founder cells produce Spitz (the Drosophila
TGFalpha homolog) signal, which is passed to the neighboring secondary Bolwig's
organ cells where it activates the EGFR signaling cascade and maintains the fate
of these secondary cells. The regulators of tll expression in the embryonic
visual system remain elusive, as we were unable to find evidence for regulation
by the 'early eye genes' so, eya and ey, or by EGFR signaling. A role for the Pax-6 homologue eyeless in adult Drosophila brain development and
function is described. eyeless expression is detected in neurons, but not glial
cells, of the mushroom bodies, the medullar cortex, the lateral horn, and the
pars intercerebralis. Furthermore, severe defects in adult brain structures
essential for vision, olfaction, and for the coordination of locomotion are
provoked by two newly isolated mutations of Pax-6/eyeless that result in
truncated proteins. Consistent with the morphological lesions, we observe
defective walking behavior for these eyeless mutants. The implications of these
data for understanding postembryonic brain development and function in
Drosophila are discussed. We analyzed the expression and function of eyeless (ey) and twin of eyeless
(toy) in the embryonic central nervous system (CNS) of Drosophila. Both genes
are differentially expressed in specific neuronal subsets (but not in glia) in
every CNS neuromere, and in the brain, specific cell populations co-expressing
both proteins define a longitudinal domain which is intercalated between broad
exclusive expression domains of ey and toy. Studies of genetic null alleles and
dsRNA interference did not reveal any gross neuroanatomical effects of ey, toy,
or ey/toy elimination in the embryonic CNS. In contrast, targeted misexpression
of ey, but not of toy, resulted in profound axonal abnormalities in the
embryonic ventral nerve cord and brain. Pax-6 genes, known to be essential for eye development, encode an evolutionarily
conserved transcription factor with two DNA-binding domains. To corroborate the
contribution of each DNA-binding domain to eye formation, we generated truncated
forms of the Drosophila Pax-6 gene eyeless and tested their capacity to rescue
the ey(2) mutant. Surprisingly, EY deleted of the homeodomain rescued the ey(2)
mutant and triggered ectopic eyes morphogenesis. In contrast, EY lacking the
paired domain failed to rescue the ey(2) mutant, led to truncation of
appendages, and repressed Distal-less when misexpressed. This result suggests
distinct functions mediated differentially by the two DNA-binding domains of
eyeless. In 1995, the eyeless (ey) gene was dubbed the "master-regulator" of eye
development in Drosophila. Not only is ey required for eye development, but its
misexpression can convert many other tissues into eye, including legs, wings and
antennae.(1) ey is remarkable for its ability to drive coordinate
differentiation of the multiple cell types that have to differentiate in a very
precise pattern to construct the fly eye, and for its power to override the
previous differentiation programs of many other diverse tissues. Even more
remarkable, the ey homolog Pax6 and homologs of other eye determination genes
from Drosophila are also required for eye development in vertebrates,(2,3)
prompting reassessment of the evolution of vision throughout the animal
kingdom.(4,5) Now Kumar and Moses have published a study that throws a new light
on ey function in Drosophila.(6) According to their work, ey becomes a master
regulator of eye development much later than previously thought, and is
regulated by signalling through the Notch and EGFR signaling pathways. Eye specification in Drosophila is thought be controlled by a set of seven
nuclear factors that includes the Pax6 homolog, Eyeless. This group of genes is
conserved throughout evolution and has been repeatedly recruited for eye
specification. Several of these genes are expressed within the developing eyes
of vertebrates and mutations in several mouse and human orthologs are the
underlying causes of retinal disease syndromes. Ectopic expression in Drosophila
of any one of these genes is capable of inducing retinal development, while
loss-of-function mutations delete the developing eye. These nuclear factors
comprise a complex regulatory network and it is thought that their combined
activities are required for the formation of the eye. We examined the expression
patterns of four eye specification genes, eyeless (ey), sine oculis (so), eyes
absent (eya), and dachshund (dac) throughout all time points of embryogenesis
and show that only eyeless is expressed within the embryonic eye anlagen. This
is consistent with a recently proposed model in which the eye primordium
acquires its competence to become retinal tissue over several time points of
development. We also compare the expression of Ey with that of a putative
antennal specifying gene Distal-less (Dll). The expression patterns described
here are quite intriguing and raise the possibility that these genes have even
earlier and wide ranging roles in establishing the head and visual field. The Drosophila compound eye is specified by the simultaneous and interdependent
activity of transcriptional regulatory genes from four families: PAX6 (eyeless,
twin of eyeless, eyegone), EYA (eyes absent), SIX (sine oculis, Optix) and DACH
(dachshund). Mammals have homologues of all these genes, and many of them are
expressed in the embryonic or adult eye, but the functional relationships
between them are currently much less clear than in Drosophila. Nevertheless,
mutations in the mammalian genes highlight their requirement both within and
outside the eye in embryos and adults, and emphasize that they can be deployed
in many different contexts. We have cloned a chick homologue of Drosophila dachshund (dac), termed Dach1.
Dach1 is the orthologue of mouse and human Dac/Dach (hereafter referred to as
Dach1). We show that chick Dach1 is expressed in a variety of sites during
embryonic development, including the eye and ear. Previous work has demonstrated
the existence of a functional network and genetic regulatory hierarchy in
Drosophila in which eyeless (ey, the Pax6 orthologue), eyes absent (eya), and
dac operate together to regulate Drosophila eye development, and that ey
regulates the expression of eya and dac. We find that in the developing eye of
both chick and mouse, expression domains of Dach1 overlap with those of Pax6, a
gene required for normal eye development. Similarly, in the developing ear of
both mouse and chick, Dach1 expression overlaps with the expression of another
Pax gene, Pax2. In the mouse, Dach1 expression in the developing ear also
overlaps with the expression of Eya1 (an eya homologue). Both Pax2 and Eya1 are
required for normal ear development. Our expression studies suggest that the
Drosophila Pax-eya-dac regulatory network may be evolutionarily conserved such
that Pax genes, Eya1, and Dach1 may function together in vertebrates to regulate
neural development. To address the further possibility that a regulatory
hierarchy exists between Pax, Eya, and Dach genes, we have examined the
expression of mouse Dach1 in Pax6, Pax2 and Eya1 mutant backgrounds. Our results
indicate that Pax6, Pax2, and Eya1 do not regulate Dach1 expression through a
simple linear hierarchy. Loss of Pax 6 function leads to an eyeless phenotype in both mammals and
insects, and ectopic expression of both the Drosophila and the mouse gene leads
to the induction of ectopic eyes in Drosophila, which suggested to us that Pax 6
might be a universal master control gene for eye morphogenesis. Here, we report
the reciprocal experiment in which the RNAs of the Drosophila Pax 6 homologs,
eyeless and twin of eyeless, are transferred into a vertebrate embryo; i.e.,
early Xenopus embryos at the 2- and 16-cell stages. In both cases, ectopic eye
structures are formed. To understand the genetic program specifying eye
morphogenesis, we have analyzed the regulatory mechanisms of Pax 6 expression
that initiates eye development. Previously, we have demonstrated that Notch
signaling regulates the expression of eyeless and twin of eyeless in Drosophila.
Here, we show that in Xenopus, activation of Notch signaling also induces
eye-related gene expression, including Pax 6, in isolated animal caps. In
Xenopus embryos, the activation of Notch signaling causes eye duplications and
proximal eye defects, which are also induced by overexpression of eyeless and
twin of eyeless. These findings indicate that the gene regulatory cascade is
similar in vertebrates and invertebrates. The two Pax6 gene homologs eyeless and twin of eyeless play decisive early roles
in Drosophila eye development. Strong mutants of twin of eyeless or of eyeless
are headless, which suggests that they are required for the development of all
structures derived from eye-antennal discs. The activity of these genes is
crucial at the very beginning of eye-antennal development in the primordia of
eye-antennal discs when eyeless is first activated by the twin of eyeless gene
product. This activation does not strictly depend on the Twin of eyeless
protein, but is temperature-dependent in its absence. Twin of eyeless acts also
in parallel to the eyeless gene and exerts functions that are partially
redundant with those of Eyeless, while Eyeless is mainly required to prevent
early cell death and promote eye development in eye-antennal discs. eyeless (ey) is a key regulator of the eye development pathway in Drosophila.
Ectopic expression of ey can induce the expression of several eye-specification
genes (eya, so, and dac) and induce eye formation in multiple locations on the
body. However, ey does not induce eye formation everywhere where it is
ectopically expressed, suggesting that EY needs to collaborate with additional
factors for eye induction. We examined ectopic eye induction by EY in the wing
disc and found that eye induction was spatially restricted to the posterior
compartment and the anterior-posterior (A/P) compartmental border, suggesting a
requirement for both HH and DPP signaling. Although EY in the anterior
compartment induced dpp and dac, these were not sufficient for eye induction.
Coexpression experiments show that EY needs to collaborate with high level of HH
and DPP to induce ectopic eye formation. Ectopic eye formation also requires the
activation of an eye-specific enhancer of the endogenous hh gene. Pax6 genes encode transcription factors with two DNA-binding domains that are
highly conserved during evolution. In Drosophila, two Pax6 genes function in a
pathway in which twin of eyeless (toy) directly regulates eyeless (ey), which is
necessary for initiating the eye developmental pathway. To investigate the gene
duplication of Pax6 that occurred in holometabolous insects like Drosophila and
silkworm, we used different truncated forms of toy and small eyes (sey), and
tested their capacity to induce ectopic eye development in an ey-independent
manner. Even though the Paired domains of TOY and SEY have DNA-binding
properties that differ from those of the Paired domain of EY, they all are
capable of inducing ectopic eye development in an ey mutant background. We also
show that one of the main functional differences between toy and ey lies in the
C-terminal region of their protein products, implying differences in their
transactivation potential. Furthermore, we show that only the homeodomain (HD)
of EY is able to downregulate the expression of Distal-less (Dll), a feature
that is required during endogenous eye development. These results suggest
distinct functions of the two DNA-binding domains of TOY and EY, and significant
evolutionary divergence between the two Drosophila Pax6 genes. Pax genes encode DNA binding proteins that play pivotal roles in the
determination of complex tissues. Members of one subclass, Pax6, function as
selector genes and play key roles in the retinal development of all seeing
animals. Mutations within the Pax6 homologs including fly eyeless, mouse Small
eye and human Pax6 lead to severe retinal defects in their respective systems.
In Drosophila eyeless and twin of eyeless, play non-redundant roles in the
developing retina. One particularly interesting characteristic of these genes is
that, although expression of either gene can induce ectopic eye formation in
non-retinal tissues, there are differences in the location and frequencies at
which the eyes develop. eyeless induces much larger ectopic eyes, at higher
frequencies, and in a broader range of tissues than twin of eyeless. In this
report we describe a series of experiments conducted in both yeast and flies
that has identified protein modules that are responsible for the differences in
tissue transformation. These domains appear to contain transcriptional activator
and repressor activity of distinct strengths. We propose a model in which the
selective presence of these activities and their relative strengths accounts, in
part, for the disparity to which ectopic eyes are induced in response to the
forced expression of eyeless and twin of eyeless. The identification of both
transcriptional activator and repressor activity within the Pax6 protein
furthers our understanding of how this gene family regulates tissue
determination. Pax6 genes encode evolutionarily highly conserved transcription factors that are
required for eye and brain development. Despite the characterization of
mutations in Pax6 homologs in a range of organisms, and despite functional
studies, it remains unclear what the relative importance is of the various parts
of the Pax6 protein. To address this, we have studied the Drosophila Pax6
homolog eyeless. Specifically, we have generated new eyeless alleles, each with
single missense mutations in one of the four domains of the protein. We show
that these alleles result in abnormal eye and brain development while
maintaining the OK107 eyeless GAL4 activity from which they were derived. We
performed in vivo functional rescue experiments by expressing in an
eyeless-specific pattern Eyeless proteins in which either the paired domain, the
homeodomain, or the C-terminal domain was deleted. Rescue of the eye and brain
phenotypes was only observed when full-length Eyeless was expressed, while all
deletion constructs failed to rescue. These data, along with the phenotypes
observed in the four newly characterized eyeless alleles, demonstrate the
requirement for an intact Eyeless protein for normal Drosophila eye and brain
development. They also suggest that some endogenous functions may be obscured in
ectopic expression experiments. Organ development is directed by selector gene networks. Eye development in the
fruit fly Drosophila melanogaster is driven by the highly conserved selector
gene network referred to as the "retinal determination gene network," composed
of approximately 20 factors, whose core comprises twin of eyeless (toy), eyeless
(ey), sine oculis (so), dachshund (dac), and eyes absent (eya). These genes
encode transcriptional regulators that are each necessary for normal eye
development, and sufficient to direct ectopic eye development when misexpressed.
While it is well documented that the downstream genes so, eya, and dac are
necessary not only during early growth and determination stages but also during
the differentiation phase of retinal development, it remains unknown how the
retinal determination gene network terminates its functions in determination and
begins to promote differentiation. Here, we identify a switch in the regulation
of ey by the downstream retinal determination genes, which is essential for the
transition from determination to differentiation. We found that central to the
transition is a switch from positive regulation of ey transcription to negative
regulation and that both types of regulation require so. Our results suggest a
model in which the retinal determination gene network is rewired to end the
growth and determination stage of eye development and trigger terminal
differentiation. We conclude that changes in the regulatory relationships among
members of the retinal determination gene network are a driving force for key
transitions in retinal development. Pax6 transcription factors are essential upstream regulators in the developing
anterior brain and peripheral visual system of most bilaterian animals. While a
single homolog is in charge of these functions in vertebrates, two Pax6 genes
are in Drosophila: eyeless (ey) and twin of eyeless (toy). At first glance,
their co-existence seems sufficiently explained by their differential
involvement in the specification of two types of insect visual organs: the
lateral compound eyes (ey) and the dorsal ocelli (toy). Less straightforward to
understand, however, is their genetic redundancy in promoting defined early and
late growth phases of the precursor tissue to these organs: the eye-antennal
imaginal disc. Drawing on comparative sequence, expression, and gene function
evidence, I here conclude that this gene regulatory network module dates back to
the dawn of arthropod evolution, securing the embryonic development of the
ocular head segment. Thus, ey and toy constitute a paradigm to explore the
organization and functional significance of longterm conserved genetic
redundancy of duplicated genes. Indeed, as first steps in this direction, recent
studies uncovered the shared use of binding sites in shared enhancers of target
genes that are under redundant (string) and, strikingly, even subfunctionalized
control by ey and toy (atonal). Equally significant, the evolutionarily recent
and paralog-specific function of ey to repress the transcription of the antenna
fate regulator Distal-less offers a functionally and phylogenetically
well-defined opportunity to study the reconciliation of shared, partitioned, and
newly acquired functions in a duplicated developmental gene pair. Eyeless (ey) is one of the most critical transcription factors for initiating
the entire eye development in Drosophila. However, the molecular mechanisms
through which Ey regulates target genes and pathways have not been characterized
at the genomic level. Using ChIP-Seq, we generated an endogenous Ey-binding
profile in Drosophila developing eyes. We found that Ey binding occurred more
frequently at promoter compared to non-promoter regions. Ey promoter binding was
correlated with the active transcription of genes involved in development and
transcription regulation. An integrative analysis revealed that Ey directly
regulated a broad and highly connected genetic network, including many essential
patterning pathways, and known and novel eye genes. Interestingly, we observed
that Ey could target multiple components of the same pathway, which might
enhance its control of these pathways during eye development. In addition to
protein-coding genes, we discovered Ey also targeted non-coding RNAs, which
represents a new regulatory mechanism employed by Ey. These findings suggest
that Ey could use multiple molecular mechanisms to regulate target gene
expression and pathway function, which might enable Ey to exhibit a greater
flexibility in controlling different processes during eye development. The insect central complex (CX) is a conserved brain region containing 60 +
neuronal subtypes, several of which contribute to navigation. It is not known
how CX neuronal diversity is generated or how developmental origin of subtypes
relates to function. We mapped the developmental origin of four key CX subtypes
and found that neurons with similar origin have similar axon/dendrite targeting.
Moreover, we found that the temporal transcription factor (TTF) Eyeless/Pax6
regulates the development of two recurrently-connected CX subtypes: Eyeless loss
simultaneously produces ectopic P-EN neurons with normal axon/dendrite
projections, and reduces the number of E-PG neurons. Furthermore, transient loss
of Eyeless during development impairs adult flies' capacity to perform celestial
navigation. We conclude that neurons with similar developmental origin have
similar connectivity, that Eyeless maintains equal E-PG and P-EN neuron number,
and that Eyeless is required for the development of circuits that control adult
navigation. |
Are enhancers directional in their targeting of gene promoters? | Promoters initiate transcription in opposite directions and are separated only by a short enhancer region, which is likely to regulate both promoters simultaneously. Most enhancers are able to regulate promoters on either side. | In cell lines established from Marek's disease tumors, several viral transcripts
are expressed and among them the products of pp38/pp24 mRNA and 1.8 kb-mRNA have
been suggested to be involved in viral oncogenicity. The long inverted repeats
of Marek's Disease virus serotype 1 (MDV1) genome contain closely located
transcriptional promoters for phosphorylated protein pp38/pp24 and 1.8 kb-mRNA.
These promoters initiate transcription in opposite directions and are separated
only by a short enhancer region, which is likely to regulate both promoters
simultaneously. We have analyzed the transcription activity of these promoters
in MDV1 (Md5 strain) infected CEF by transient expression of CAT reporter genes
and found that the promoters were in fact active in infected cells and the
promoter for 1.8 kb-mRNA was more active than the pp38/pp24 promoter. Deletion
analysis of the short enhancer region revealed that the 30 bp region overlapping
the enhancer elements for 1.8 kb-mRNA was important for promoter activity for
pp38/pp24. The gel shift analysis revealed that nuclear factor(s) actually bound
to the overlapping 30 bp region. In addition, the activity of these promoters in
infected cells varied with MDV strains. These results suggest that pp38/pp24 and
1.8 kb-mRNA promoters share a common regulatory sequence but a viral or a
cellular factor(s) induced by viral infection regulates the promoter by distinct
mechanisms. |
Has dupilumab been FDA approved for atopic dermatitis? | Yes, dupilumab has been approved by FDA for atopic dermatitis. | Atopic dermatitis (AD) is a chronic, inflammatory skin disease characterized by
pruritus, inflammatory erythematous skin lesions, and skin-barrier defect.
Current mainstay treatments of emollients, steroids, calcineurin inhibitors, and
immunosuppressants have limited efficacy and potentially serious side effects.
Recent advances and understanding of the pathogenesis of AD have resulted in new
therapies that target specific pathways with increased efficacy and the
potential for less systemic side effects. New FDA-approved therapies for AD are
crisaborole and dupilumab. The JAK-STAT inhibitors (baricitinib, upadacitinib,
PF-04965842, ASN002, tofacitinib, ruxolitinib, and delgocitinib) have the most
promising results of the emerging therapies. Other drugs with potential include
the aryl hydrocarbon receptor modulating agent tapinarof, the IL-4/IL-13
antagonists lebrikizumab and tralokinumab, and the IL-31Rα antagonist
nemolizumab. In this review, new and emerging AD therapies will be discussed
along with their mechanisms of action and their potential based on clinical
study data. Dupilumab inhibits the interleukin-4 receptor subunit α and is FDA approved for
treatment of moderate-to-severe atopic dermatitis. It is a relatively new drug,
and whether it is efficacious for other diseases in dermatology is an area of
increasing interest. We searched the literature and ClinicalTrials.gov database
for uses of dupilumab beyond atopic dermatitis in dermatology and for ongoing
studies on new uses for dupilumab. Off-label reports identified described use of
dupilumab for several different dermatologic conditions, including allergic
contact dermatitis, hand dermatitis, chronic spontaneous urticaria, prurigo
nodularis, and alopecia areata. Overall, there is limited but promising data for
dupilumab use beyond atopic dermatitis in dermatology. The relatively safe
adverse effect profile of dupilumab may make it an option for certain
recalcitrant diseases in dermatology, but further studies will be needed to
assess its efficacy and determine its best possible use. J Drugs Dermatol.
2019;18(10):1053-1055. Dupilumab is the first US FDA approved biologic for treatment of atopic
dermatitis. It is a human monoclonal antibody which blocks the shared receptor
component, the interleukin (IL)-4α subunit, of IL-4 and IL-13 signaling
pathways. Occurrence of "conjunctivitis", mostly in atopic dermatitis trials,
has been the main side effect reported thus far. The etiology of
"conjunctivitis" associated with dupilumab treatment is unclear and might be
similar to atopic keratoconjunctivitis. There is evidence in the published
literature that unlike the Th2-like profile in vernal keratoconjunctivitis,
Th1-mediated inflammation is predomit in atopic keratoconjunctivitis.
Blocking the Th2 pathway with dupilumab therapy might result in a shift towards
Th1, causing the ocular findings associated with dupilumab. In addition,
blockage of IL-13 might have implications with regards to mucin production and
ocular surface health. This review highlights the clinical manifestations,
reviews treatment options and offers explanations for pathogenesis of this
ocular surface diseases associated with dupilumab treatment. |
Describe ReactomeGSA | ReactiveomeGSA is a novel resource for comparative pathway analyses of multi-omics datasets. Data from different species is automatically mapped to a common pathway space. Public data from ExpressionAt Atlas and Single Cell ExpressionAtlas can be directly integrated in the analysis. reactomegSA greatly reduces the technical barrier for multi-CSF, cross-species, and Comparative Pathways analysis. | Pathway analyses are key methods to analyze 'omics experiments. Nevertheless,
integrating data from different 'omics technologies and different species still
requires considerable bioinformatics knowledge.Here we present the novel
ReactomeGSA resource for comparative pathway analyses of multi-omics datasets.
ReactomeGSA can be used through Reactome's existing web interface and the novel
ReactomeGSA R Bioconductor package with explicit support for scRNA-seq data.
Data from different species is automatically mapped to a common pathway space.
Public data from ExpressionAtlas and Single Cell ExpressionAtlas can be directly
integrated in the analysis. ReactomeGSA greatly reduces the technical barrier
for multi-omics, cross-species, comparative pathway analyses.We used ReactomeGSA
to characterize the role of B cells in anti-tumor immunity. We compared B cell
rich and poor human cancer samples from five of the Cancer Genome Atlas (TCGA)
transcriptomics and two of the Clinical Proteomic Tumor Analysis Consortium
(CPTAC) proteomics studies. B cell-rich lung adenocarcinoma samples lacked the
otherwise present activation through NFkappaB. This may be linked to the
presence of a specific subset of tumor associated IgG+ plasma cells that lack
NFkappaB activation in scRNA-seq data from human melanoma. This showcases how
ReactomeGSA can derive novel biomedical insights by integrating large
multi-omics datasets. |
What is the role of phenylbutyrate–taurursodiol for amyotrophic lateral sclerosis? | Treatment of amyotrophic lateral sclerosis patients with phenylbutyrate–taurursodiol was associated with both functional and survival benefits. | Conflict of interest statement: S. Paganoni reports grants from Amylyx
Pharmaceuticals, Inc, The ALS Association, and ALS Finding A Cure during the
conduct of the study, and grants from Revalesio, Ra Pharma, Biohaven, Clene
Nanomedicine, and Prilenia, outside the submitted work. S. Hendrix reports other
personal fees from Pentara Corporation, outside the submitted work. S.P. Dickson
reports other personal fees from Pentara outside the submitted work. E.A.
Macklin reports grants from Amylyx, The ALS Association, and ALS Finding A Cure
Foundation during the conduct of the study. He is also a data and safety
monitoring board member (DSMB) and reports grants from Acorda Therapeutics;
steering committee membership for Biogen; consultant work for Cerevance; grants
from GlaxoSmithKline; consultant work for Inventram, Lavin Consulting, and
Myolex; grants from Mitsubishi Tanabe Pharmaceuticals; and DSMB membership for
Novartis Pharmaceuticals and Shire Human Genetic Therapies, outside the
submitted work. J.D. Berry reports grants from ALS Finding A Cure, The ALS
Association, and Amylyx during the conduct of the study; personal fees from
Biogen and Clene Nanomedicine; and grants from Alexion, Biogen, MT Pharma of
America, Anelixis Therapeutics, Brainstorm Cell Therapeutics, Genentech, nQ
Medical, National Institute of Neurological Disorders and Stroke, and the
Muscular Dystrophy Association, outside the submitted work. M.A. Elliott has
received personal fees from Amylyx and personal fees from Biogen. C. Karam
reports grants and personal fees from Akcea, Alnylam, and Genzyme, and personal
fees from Acceleron, Biogen, Alexion, Argenx, Cytokinetics, and CSL Behring,
outside the submitted work. J.B. Caress reports grants from Amylyx during the
conduct of the study, and grants from Orion Pharmaceuticals, MTB Pharma, and
Cytokinetics, outside the submitted work. J. Wymer reports grants from Amylyx
during the conduct of the study. S.A. Goutman reports grants from The ALS
Association during the conduct of the study; grants from the National Institutes
of Health/National Institutes of Environmental Health Sciences, The ALS
Association, and Target ALS, outside the submitted work; consultant work for
Biogen and ITF Pharma, outside the submitted work; and personal fees from
Biogen, ITF Pharma, Watermark Research Partners, and for expert testimony,
outside the submitted work. T.D. Heiman‐Patterson reports grants from Mitsubishi
Tanabe Pharma America, Amylyx Pharmaceuticals, The ALS Association, and Orion
Pharma, and personal fees from Cytokinetics, ITF, and Biohaven, outside the
submitted work. C.E. Jackson reports grants from Amylyx during the conduct of
the study; grants and personal fees from Cytokinetics, personal fees from CSL
Behring, grants and personal fees from Mitsubishi Tanabe Pharma America, DSMB
membership, and personal fees from Brainstorm and Mallinckrodt during the
conduct of this study; and personal fees from ITF Pharma, outside the submitted
work. C. Quinn received personal fees for serving on an advisory board of
Amylyx, outside the submitted work. J.D. Rothstein reports licensing agreement
and nonficial support from Ionis Pharmaceuticals; nonficial support from
Calico, Biogen, and IBM Watson; research grant support from the National
Institute of Neurological Disorders and Stroke, National Institute on Aging,
Department of Defense, the Chan Zuckerberg Initiative, Microsoft, The ALS
Association, the Muscular Dystrophy Association, Target ALS, F Prime, ALS
Finding A Cure, Answer ALS, Robert Packard Center for ALS Research,
GlaxoSmithKline, Travelers Insurance, American Airlines, Caterpillar, and the
National Football League; and personal consulting fees from Expansion
Therapeutics and Team Gleason. He also reports that his institution was a trial
site and thus had a contract with Amylyx to participate in the study. J. Katz
reports personal fees from MT Pharma America, Denali Pharmaceuticals, Genentech,
and Calico, outside the submitted work. S. Ladha reports grants from Amylyx,
Biogen, and MT Pharma, and personal fees from Amylyx and Biogen. T.M. Miller
reports licensing agreement and nonficial support from Ionis Pharmaceuticals,
a licensing agreement with C2N, grants and personal fees from Biogen, and
personal fees from Cytokinetics and Disarm Therapeutics, outside the submitted
work. S.N. Scelsa reports grants from Amylyx during the conduct of the study,
and grants from Orion Pharma, outside the submitted work. T.H. Vu reports
personal fees from the speakers bureau of Mitsubishi Tanabe Pharmaceuticals, and
has participated in clinical trials sponsored by Amylyx, Orion, Biogen,
Mallinckrodt, and Cytokinetics during the conduct of the study. J.D. Glass
reports that his institution was a trial site and thus had a contract from
Amylyx to participate in the study. A. Swenson reports research support from
Amylyx, The ALS Association, Massachusetts General Hospital, the National
Institutes of Health/National Institute of Neurological Disorders and Stroke,
and serving on an independent data monitoring committee for Alexion. P.L. Andres
reports personal fees from Amylyx for consulting during the conduct of the study
and has an isometric strength testing apparatus (US Patent 7493812B2) held by
the Hospital Corporation. S. Babu reports research support from the American
Academy of Neurology, the AANEM Foundation, The ALS Association, the Muscular
Dystrophy Association, Biogen, Orion, Voyager Therapeutics, and Novartis. M.
Chase reports grants to the Massachusetts General Hospital (MGH) from The ALS
Association, grants to the MGH from ALS Finding A Cure, and fee for service from
Amylyx during the conduct of the study. M. Hall reports grants for funding for
clinical trial monitoring and outcomes training support from The ALS Association
and Amylyx during the conduct of the study. G. Kittle reports grants from The
ALS Association and Amylyx during the conduct of the study. J.M. Shefner reports
grants and personal fees from Amylyx during the conduct of the study; personal
fees for consulting work from Cytokinetics and Brainstorm; grants and personal
fees for outcomes training and study design from Mitsubishi Pharma America,
outside the submitted work; personal fees for consulting from Neurosense and
Otsuka, outside the submitted work; and grants for outcomes training from
Alexion, Medicinova, and Biogen, outside the submitted work. He is also
Neuromuscular section editor for UpToDate. J. Wittes and Z.‐F. Yu report
payments from Amylyx to their employer during the conduct of the study. J. Cohen
and J. Klee report a relationship with Amylyx during the conduct of the study,
and they serve as co‐CEOs of Amylyx, outside the submitted work, with multiple
patents issued to Amylyx. K. Leslie reports being full‐time employee of Amylyx
during the conduct of the study, and personal fees from Amylyx, outside the
submitted work. R.E. Tanzi reports personal fees from Amylyx outside the
submitted work; has helped with inception and design of the clinical trial but
was not involved with running the trial and had no contact with the trial
subjects; and owns founding equity in Amylyx and serves as head of the company's
scientific advisory board. W. Gilbert was director of Amylyx during the conduct
of the study and a company shareholder. P.D. Yeramian reports full‐time
employment at Amylyx during the conduct of the study. D. Schoenfeld reports
grants from The ALS Association, during the conduct of the study, and personal
fees from Immunitypharma and Alexion, outside the submitted work. M.E. Cudkowicz
reports grants from Massachusetts General Hospital during the conduct of the
study; grants from Clene Nanomedicine, Ra Pharma, Biohaven, and Prilenia,
outside the submitted work; and personal fees from Takeda, Biogen, Sunovian,
Cytokinetics, and Immunity Pharma, outside the submitted work. The remaining
authors declare no potential conflicts of interest. |
What is known about growth arrest-specific 6 protein? | Growth arrest-specific 6 (Gas6) is a vitamin K-dependent protein secreted by immune cells, endothelial cells, vascular smooth muscle cells, and adipocytes. Recent studies indicate that Gas6 and receptors of the TAM (Tyro3, Axl, and Mer) family may be involved in the pathogenesis of obesity, systemic inflammation, and insulin resistance. | BACKGROUND: Growth arrest-specific 6 (Gas6) is a vitamin K-dependent protein
secreted by immune cells, endothelial cells, vascular smooth muscle cells, and
adipocytes. Recent studies indicate that Gas6 and receptors of the TAM (Tyro3,
Axl, and Mer) family may be involved in the pathogenesis of obesity, systemic
inflammation, and insulin resistance. The aim of this study was to investigate
the association between plasma Gas6 protein and the c.843 + 7G>A Gas6
polymorphism in metabolic syndrome (MetS).
METHODS: Two hundred five adults (88 men and 117 women) were recruited in this
study. Plasma Gas6 concentration, general, and biochemical data were measured.
All subjects were genotyped for the c.843 + 7G>A Gas6 polymorphism.
RESULTS: Plasma Gas6 concentrations decreased in parallel with various MetS
components in all groups (P = 0.017 for trend). Patients in the second and third
tertiles of Gas6 level had higher high-density lipoprotein cholesterol (HDL-C)
levels than those in the first tertile overall and in the female group. Plasma
Gas6 levels were significantly positively correlated with HDL-C level and
negatively with fasting glucose level in the female patients. The A allele and
genotype AA in single nucleotide polymorphism c.843 + 7G>A were less frequent in
the subjects with MetS compared to those without MetS.
CONCLUSIONS: Our results demonstrated a positive correlation between Gas6
protein values and HDL-C and reinforce the association with fasting glucose. In
addition, the presence of c.843 + 7G>A Gas6 polymorphisms, especially the AA
genotype, had an association with MetS. The potential role of the Gas6/TAM
system in MetS deserves further investigation. AIMS/INTRODUCTION: Obesity is characterized by disturbed adipocytokine
expression and insulin resistance in adipocytes. Growth arrest-specific 6 (GAS6)
is a gene encoding the Gas6 protein, which is expressed in fibroblasts, and its
related signaling might be associated with adipose tissue inflammation, glucose
intolerance and insulin resistance. The aim of this study was to investigate the
associations among Gas6, adipocytokines and insulin resistance in adipocytes.
MATERIALS AND METHODS: Mature Simpson Golabi Behmel Syndrome adipocytes were
treated with high levels of insulin to mimic insulin resistance, and were
examined for the expressions of Gas6, cytokines and adipocytokines from
preadipocytes in differentiation. In an animal study, high-fat diet-induced
obese mice were used to verify the Gas6 expression in vitro.
RESULTS: During the differentiation of adipocytes, the expression of Gas6
gradually decreased, and was obviously downregulated with adipocyte inflammation
and insulin resistance. Gas6 levels were found to be in proportion to the
expression of adiponectin, which has been regarded as closely relevant to
improved insulin sensitivity after metformin treatment. Similar results were
also confirmed in the animal study.
CONCLUSIONS: Our results suggest that Gas6 might modulate the expression of
adiponectin, and might therefore be associated with insulin resistance in
adipose tissues. |
What is Leptomeningeal disease? | Neoplastic leptomeningeal disease (LMD) represents infiltration of the leptomeninges by tumor cells. | |
What percent of Rheumatoid Arthritis (RA) patients are not responding to anti-TNF therapy? | These therapies are, however, expensive and 30% of patients fail to respond. | BACKGROUND: Anti-tumour necrosis factor-alpha (TNF-alpha) therapies represent an
important advancement in therapy for rheumatoid arthritis (RA). However, there
remains a proportion of patients who do not improve despite therapy. These drugs
are expensive and have the potential of serious toxicity. Therefore, it would be
ideal to predict the patients who will respond, so that the use of these drugs
can be targetted.
OBJECTIVE: To identify the clinical factors present at the start of
anti-TNF-alpha therapy that are associated with response at 6 months in patients
with RA.
METHODS: The British Society for Rheumatology (BSR) Biologics Register collects
detailed data on all patients with a rheumatic disease receiving biologic
therapy in the UK. We studied all patients with RA who had started etanercept
(ETA) or infliximab (INF) and had achieved a minimum 6 months follow-up by 1
October, 2004. The disease status at the baseline and at 6 months was assessed
using the Disease Activity Score (DAS28). The response was classified according
to the European League against Rheumatism (EULAR) improvement criteria. The
effect of baseline characteristics on response was studied using multivariate
ordinal logistic regression.
RESULTS: 2879 patients were included in this analysis (1267 ETA, 1612 INF). At
the start of therapy, the mean age was 55 yrs, disease duration 14 yrs, baseline
DAS28 6.7 and health assessment questionnaire (HAQ) 2.1. In all, 28% of ETA and
86% of INF patients were receiving methotrexate. After 6 months, 18% had a good
EULAR response, of whom 9% were considered to be in remission and 50% had a
moderate response. There was no overall difference in response rate between the
two anti-TNF-alpha therapies. A higher baseline HAQ score correlated with a
lower response rate while a better response was associated with the current use
of NSAIDs and the use of methotrexate (MTX), although the latter only reached
statistical significance with ETA [OR 1.82 (95% CI 1.38-2.40)]. There was a
lower response rate among current smokers, particularly in patients receiving
INF [OR 0.77 (95% CI 0.60-0.99)]. Age, disease duration, rheumatoid factor and
the previous number of disease-modifying antirheumatic drugs (DMARDs) did not
predict response to either drug. However, females were less likely to achieve
remission.
CONCLUSIONS: These data support an improved outcome among patients receiving MTX
in combination with anti-TNF-alpha therapies. However, the most disabled
patients were less likely to respond, despite concurrent MTX. The benefits of
NSAIDs may reflect the relative absence of comorbidities in patients who can
tolerate these drugs or the continuing presence of reversible inflammatory
symptoms. The association of smoking and poor outcome with INF is a novel
finding and may reflect alterations in pharmacokinetics. The inability of other
baseline disease characteristics to predict the outcome suggests that other
factors, including potential genetic differences in drug metabolism, may be
influencing the response to anti-TNF-alpha therapies. Anti-TNF treatments have given patients with rheumatoid arthritis considerable
hope and relief. However, 20-30% of patients do not respond sufficiently to a
given anti-TNF drug. In this situation, current strategies include switching to
an alternative agent, increasing the dose of the current agent or to return to
conventional DMARDs. The arrival of new biologics, which target different
molecules than TNF, opens the perspective to other pathways of immunomodulation
in RA. These drugs include rituximab (anti-CD20), abatacept (CTLA4Ig) and
tocilizumab (anti- IL6R). Comparative studies are urgently needed to assess the
efficacy of the different treatment options in order to provide the best
therapeutic strategy for our patients. OBJECTIVE: To evaluate the effectiveness and safety of anti-tumor necrosis
factor (anti-TNF) therapies in rheumatoid arthritis (RA), and to identify the
factors involved in this response.
METHODS: Dynamic prospective cohort study of patients with RA treated with
anti-TNF under clinical practice conditions. Effectiveness was evaluated using
Disease Activity Score (DAS) 28, European League Against Rheumatism (EULAR)
response, Health Assessment Questionnaire (HAQ), and time to treatment failure.
Prior adherence was evaluated retrospectively and safety was evaluated by
adverse events (AE). The analysis was restricted to anti-TNF-naive patients.
RESULTS: The study included 161 patients treated for RA during 6 years (60
infliximab, 79 etanercept, and 22 adalimumab). At 6 months, 15% reached a good
EULAR response and 38% a moderate response. A mean decrease of -1.5 (p < 0.0001)
was observed in the DAS28 and of -0.34 in the HAQ (p < 0.0001); however, women
showed poorer progress in terms of DAS and HAQ. In the first year, 64.3% did not
experience treatment failure and this figure was 50.5% after 2 years. In
one-third, glucocorticoids were withdrawn and in the remainder the dose was
reduced by 50%. Adherence to treatment, selection of etanercept, and
intensification of infliximab were associated with a lower probability of
premature failure in the multivariate model. AE were similar to other those in
studies and no outstanding differences in safety were found between the 3
anti-TNF therapies.
CONCLUSIONS: Anti-TNF treatments are effective and safe, reducing the activity
of the disease, disability, and the need for corticosteroids. Patients who
displayed good adherence prior to the anti-TNF treatment and were treated with
etanercept or with increasing doses of infliximab had the best chance of
displaying a response. OBJECTIVE: To identify the clinical factors predicting failure or a good
clinical response in the cohort of RA patients entered in the Lombardy
Rheumatology Network (LORHEN) registry after 3 years of treatment with anti-TNF
agents.
METHODS: We studied the patients who had received anti-TNF agents and been
followed up for a minimum of 6 months. Disease activity at baseline and after 6
months was assessed using the DAS28, and response was evaluated according to the
EULAR improvement criteria.
RESULTS: 1005 patients (55.72 years) were included in the analysis. at baseline
the DAS-28 was 5.91+/-0.95 and a HAQ score was 1.46+/-0.61. At mean of 14.57
months, 29.9% of the patients achieved a DAS-28 of <or=2.6 (remission). A higher
RR for remission was associated with male gender (AHR 1.51, 95% CI 1.14-2.00; p:
0.004) and a lower RR for remission with: prior treatment with >3 DMARDs (AHR
0.077, 95% CI 0.58-1.03; p: 0.074), a high ESR (AHR 0.86, 95% CI 0.81-0.92; p:
0.000), Steinbrocker's functional class III/IV (AHR 0.66, 95% CI 0.48-0.90; p:
0.010), a high TJC (AHR 0.97, 95% CI 0.94-0.99; p: 0.011). A 12-month EULAR
non-response was observed in 153/821 (18.6%) associated with a higher baseline
HAQ score (AOR 1.51, 95% CI 1.03-2.20, p: 0.033), prior treatment with >3 DMARDs
(AOR 1.76, 95% CI 1.09-2.85; p: 0.021) and corticosteroid >5 mg/day (AOR 2.05,
95% CI 1.06-3.97; p: 0.034).
CONCLUSION: We found that only a minority of patients with long-standing RA
treated with anti-TNF agents achieve a good clinical response or remission. Although TNF inhibitors have dramatically improved the outcome of patients with
rheumatoid arthritis, 30-40% of patients do not respond well to them and
treatment needs to be changed. In an effort to discriminate good and poor
responders, we focused on the change in serum and synovial fluid levels of
interleukin (IL-) 33 before and after treatment with TNF inhibitors. They were
also measured in synovial fluids from 17 TNF inhibitor-naïve patients, and
fibroblast-like synoviocytes (FLS) in-culture from 6 patients and correlated
with various pro-inflammatory cytokines. Serum levels of IL-33 at 6 months after
treatment decreased significantly in responders, while they did not change in
non-responders. Synovial fluid levels of IL-33 in 6 patients under treatment
with TNF inhibitors stayed high in 3 who were refractory and slightly elevated
in 2 moderate responders, while they were undetectable in one patient under
remission. Among inflammatory cytokines measured in 17 synovial fluids from TNF
inhibitor-naïve patients, levels of IL-33 showed a significant positive
correlation only to those of IL-1β. IL-1β increased IL-33 expression markedly in
FLS in vitro, compared to TNF-α. IL-1β might be inducing RA inflammation through
producing pro-inflammatory IL-33 in TNF inhibitor-hypo-responders. Sustained
elevation of serum and/or synovial levels of IL-33 may account for a poor
response to TNF inhibitors, although how TNF inhibitors affect the level of
IL-33 remains to be elucidated. Approximately 30% of rheumatoid arthritis patients achieve inadequate response
to anti-TNF biologics. Attempts to identify molecular biomarkers predicting
response have met with mixed success. This may be attributable, in part, to the
variable and subjective disease assessment endpoints with large placebo effects
typically used to classify patient response. Sixty-one patients with active RA
despite methotrexate treatment, and with MRI-documented synovitis, were
randomized to receive infliximab or placebo. Blood was collected at baseline and
genome-wide transcription in whole blood was measured using microarrays. The
primary endpoint in this study was determined by measuring the transfer rate
constant (Ktrans) of a gadolinium-based contrast agent from plasma to synovium
using MRI. Secondary endpoints included repeated clinical assessments with
DAS28(CRP), and assessments of osteitis and synovitis by the RAMRIS method.
Infliximab showed greater decrease from baseline in DCE-MRI Ktrans of wrist and
MCP at all visits compared with placebo (P<0.001). Statistical analysis was
performed to identify genes associated with treatment-specific 14-week change in
Ktrans. The 256 genes identified were used to derive a gene signature score by
averaging their log expression within each patient. The resulting score
correlated with improvement of Ktrans in infliximab-treated patients and with
deterioration of Ktrans in placebo-treated subjects. Poor responders showed high
expression of activated B-cell genes whereas good responders exhibited a gene
expression pattern consistent with mobilization of neutrophils and monocytes and
high levels of reticulated platelets. This gene signature was significantly
associated with clinical response in two previously published whole blood gene
expression studies using anti-TNF therapies. These data provide support for the
hypothesis that anti-TNF inadequate responders comprise a distinct molecular
subtype of RA characterized by differences in pre-treatment blood mRNA
expression. They also highlight the importance of placebo controls and robust,
objective endpoints in biomarker discovery.
TRIAL REGISTRATION: ClinicalTrials.gov NCT01313520. Although anti-TNF drugs have changed the clinical course of rheumatoid arthritis
(RA), survival rates and resistance-to-therapy data confirm that about 30% of RA
patients fail to respond. The aim of this study was to evaluate the correlations
between the development of antidrug antibodies, specific IgG4 antibodies against
TNF inhibitors, and resistance to therapy in RA patients. This retrospective
study involved 129 patients with established RA naïve to biological agents (98
females and 32 males, mean age 56.7±12.3 years, disease duration 6.3±1.2 years,
baseline Disease Activity Score [DAS]-28 3.2-5.6) who received treatment with
anti-TNF agents after the failure of conventional disease-modifying
antirheumatic drugs (32 received infliximab [IFX], 58 etanercept [ETN], and 39
adalimumab [ADA]). After 6 months of treatment, the patients were classified as
being in remission (DAS28 <2.6), having low disease activity (LDA; DAS28
2.6-3.2), or not responding (NR: DAS28 >3.2). The patients were also tested for
serum antidrug antibodies and IgG4 antibodies against TNF inhibitors. After 24
weeks of treatment, 38% of the ETN-treated patients and 28% of those treated
with ADA had injection-site reactions; the rate of systemic reactions in the IFX
group was 25%. The differences among the three groups were not statistically
significant (P=0.382; ETN versus ADA P=0.319). The percentages of patients with
adverse events stratified by drug response were: LDA 8% and NR 18% in the ADA
group; in remission 3%, LDA 22%, and NR 10% in the ETN group; and LDA 6% and NR
16% in the IFX group (P=0.051). The percentages of patients with antidrug
antibodies were: ADA 33.3%, ETN 11.5%, and IFX 10.3% (P=0.025; ADA versus ETN
P=0.015). The percentages of patients with IgG4 antibodies were: ADA 6%, ETN
13%, and IFX 26% (P=0.017; ADA versus ETN P=0.437). Associations between
antidrug antibodies, specific IgG4 antibodies, and adverse reactions were not
significant for any of the three drugs. IgG4 levels were higher in the ADA group
than in the other two groups, and higher in the patients with worse DAS28 (NR)
and in those experiencing adverse events. These data suggest a possible
association between IgG4 levels and worse DAS28 (r (2)=5.8%, P=0.011). The
presence of specific IgG4 antibodies against TNF blockers in patients with RA
might affect the drugs' activity. Patients with injection-site reactions and
IgG4 against ETN may show a decreased response. OBJECTIVES: Rheumatoid arthritis (RA) patients with moderate disease activity
show progression of joint damage and have impaired quality of life, physical
function, work and daily activities. Little is known about management of
patients with moderate RA. The aim of the study was to assess the 1-year
response to anti-TNF in biologic-naïve RA patients with moderate (3.2 <DAS28
≤5.1) disease activity despite DMARD treatment, in the Italian clinical
practice.
METHODS: The MODERATE study is a multicentre prospective, cohort
non-interventional study, conducted in 19 Italian rheumatology sites. Patients
with moderate RA, diagnosed according to the 2010 American College of
Rheumatology (ACR)/EULAR criteria, were enrolled if they also were aged ≥18
years, had disease onset after 16 years old, moderate disease at baseline (DAS28
score >3.2 and ≤5.1), and were naïve to anti-TNF treatment.
RESULTS: Among 157 RA patients, 93 (59%) underwent etanercept, 43 (22%)
adalimumab, 26 (17%) certolizumab, 10 golimumab and 2 infliximab; 80% of
patients were still in treatment after 12-month observation. One-year clinical
remission was achieved by 27 RA patients (21%), reduction of DAS28 score greater
than 1.2 was observed in 75 (58%) patients. Moderate and good response according
to EULAR criteria was observed in 59 (46%) and 45 (35%) patients, respectively.
CONCLUSIONS: Results confirm the efficacy of anti-TNF alpha also in moderate RA
patients, who may achieve a substantial decrease of disease activity, and
improve their quality of life. The low rate of patients achieving remission may
suggest that therapeutic strategies should be more timely and aggressive. |
Which recombinant antibody therapeutics were granted marketing approval in 2014? | Six recombinant antibody therapeutics (vedolizumab, siltuximab, ramucirumab, pembrolizumab, nivolumab, blinatumomab) were granted their first marketing approvals in 2014. | The commercial pipeline of recombit antibody therapeutics is robust and
dynamic. As of early December 2014, a total of 6 such products (vedolizumab,
siltuximab, ramucirumab, pembrolizumab, nivolumab, blinatumomab) were granted
first marketing approvals in 2014. As discussed in this perspective on
antibodies in late-stage development, the outlook for additional approvals,
potentially still in 2014 and certainly in 2015, is excellent as marketing
applications for 7 antibody therapeutics (secukinumab, evolocumab, mepolizumab,
dinutuximab, nivolumab, blinatumomab, necitumumab) are undergoing a first
regulatory review in the EU or US. Of the 39 novel mAbs currently in Phase 3
studies, a marketing application for one (alirocumab) may be submitted in late
2014, and marketing application submissions for at least 4 (reslizumab,
ixekizumab, ocrelizumab, obiltoxaximab) are expected in 2015. Other 'antibodies
to watch' are those in Phase 3 studies with estimated primary completion dates
in late 2014 or 2015, which includes 13 for non-cancer indications (brodalumab,
bimagrumab, bococizumab, MABp1, gevokizumab, dupilumab, sirukumab, sarilumab,
tildrakizumab, guselkumab, epratuzumab, combination of actoxumab + bezlotoxumab,
romosozumab) and 2 (racotumomab and clivatuzumab tetraxetan) undergoing
evaluation as treatments for cancer. In addition to the novel antibody
therapeutics mentioned, biosimilar infliximab and biosimilar trastuzumab are
'antibodies to watch' in 2015 because of their potential for entry into the US
market and regulatory review, respectively. |
What is caused by BACH2-related immunodeficiency? | Affected subjects of a syndrome of BACH2-related immunodeficiency and autoimmunity (BRIDA) had lymphocyte-maturation defects that caused immunoglobulin deficiency and intestinal inflammation. The mutations disrupted protein stability by interfering with homodimerization or by causing aggregation. | Author information:
(1)Lymphocyte Cell Biology Section (Molecular Immunology and Inflammation
Branch), Biodata Mining and Discovery Section and Protein Expression Laboratory,
National Institute of Arthritis and Musculoskeletal and Skin Diseases, National
Institutes of Health, Bethesda, Maryland, USA.
(2)MRC Centre for Transplantation, King's College London, London, UK.
(3)Molecular Development of the Immune System Section, NIAID Clinical Genomics
Program, Biological Imaging Section (Research Technologies Branch) and Mucosal
Immunity Section, National Institute of Allergy and Infectious Diseases,
National Institutes of Health, Bethesda, Maryland, USA.
(4)Molecular Neuroscience, Institute of Neurology, Faculty of Brain Sciences,
University College London, London, UK.
(5)Department of Medicine, Imperial College London, London, UK.
(6)Laboratory of Lymphocyte Signaling and Development, Babraham Institute,
Cambridge, UK.
(7)Translational Gastroenterology Unit, Nuffield Department of Medicine, John
Radcliffe Hospital, Oxford, UK.
(8)Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics,
Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK.
(9)Department of Biochemistry and Department of Computer Science, Purdue
University, West Lafayette, Indiana, USA.
(10)Imperial BRC Genomics Facility, Hammersmith Hospital, London, UK.
(11)Merck Research Laboratories, Merck &Co. Inc., Boston, Massachusetts, USA.
(12)Clinical Research Directorate/CMRP, Leidos Biomedical Research Inc., NCI at
Frederick, Frederick, Maryland, USA.
(13)National Cancer Institute, National Institutes of Health, Bethesda,
Maryland, USA.
(14)Department of Paediatrics, University of Oxford, Oxford, UK.
(15)Centre for Genomic and Experimental Medicine, Institute of Genetics and
Molecular Medicine, University of Edinburgh, Edinburgh, UK.
(16)Department of Haematology, Northern Centre for Cancer Care, Newcastle upon
Tyne, UK. |
Should cerebrolysin be used for aneurysmal subarachnoid hemorrhage? | No. Randomized clinical trial did not find any superior effects of cerebrolysin for patients with aneurysmal subarachnoid hemorrhage. | INTRODUCTION: Cerebrolysin is a neuroprotective drug used in the treatment of
acute ischemic stroke. To our knowledge, this drug has never been evaluated in
patients with aneurysmal subarachnoid hemorrhage (SAH). The aim of this study
was to evaluate the effect of Cerebrolysin in patients with aneurysmal SAH.
METHODS: Aneurysmal SAH patients who had their aneurysm obliterated at our
institution from 2007 to 2016 were retrospectively studied. Patients received
Cerebrolysin treatment or standard care only (control group). Subgroup analyses
were performed according to Hunt and Hess grade (good grade ≤ 2, N = 216; poor
grade ≥ 3, N = 246) and treatment procedure (clip or coil).
RESULTS: In good-grade patients (N = 216), clinical outcomes and mortality did
not differ significantly between the control and Cerebrolysin groups. In
poor-grade patients (N = 246), the mortality rate was significantly lower in the
Cerebrolysin group (8.7%) than in the control group (25.4%, p = 0.006). In
patients who received microsurgical clipping (N = 328), the mortality rate was
significantly lower in the Cerebrolysin group (7.3%) than in the control group
(18.5%, p = 0.016).
CONCLUSION: Cerebrolysin injection during the acute period of SAH appeared to
reduce the mortality rate, especially in poor-grade patients. This study
suggests the potential of Cerebrolysin for treating aneurysmal SAH. Further
studies are needed to confirm our results. |
Do bacteria release extracellular vesicles? | Yes, Bacterial extracellular vesicles (EVs) are bilayered lipid membrane structures, bearing integral proteins and able to carry diverse cargo outside the cell to distant sites. | Bacterial extracellular vesicles (EVs) are bilayered lipid membrane structures,
bearing integral proteins and able to carry diverse cargo outside the cell to
distant sites. In microorganisms, EVs carry several types of molecules:
proteins, glycoproteins, mRNAs and small RNA species, as mammalian EVs do, but
also carbohydrates. Studying EVs opens a whole new world of possibilities to
better understand the interplay between host and bacteria crosstalks, although
there are still many questions to be answered in the field, especially when it
comes to microbiota-derived EVs. In this review, we propose to summarize and
analyse the current literature about bacterial EVs and possible clinical
applications, through answering three main questions: (a) What are bacterial
EVs? (b) What are EV impacts on skin inflammatory disease physiopathology? (iii)
What are the possible and expected clinical applications of EVs to treat
inflammatory skin diseases? Gram-negative and Gram-positive bacteria release a variety of membrane vesicles
through different formation routes. Knowledge of the structure, molecular cargo
and function of bacterial extracellular vesicles (BEVs) is primarily obtained
from bacteria cultured in laboratory conditions. BEVs in human body fluids have
been less thoroughly investigated most probably due to the methodological
challenges in separating BEVs from their matrix and host-derived eukaryotic
extracellular vesicles (EEVs) such as exosomes and microvesicles. Here, we
present a step-by-step procedure to separate and characterize BEVs from human
body fluids. BEVs are separated through the orthogonal implementation of
ultrafiltration, size-exclusion chromatography (SEC) and density-gradient
centrifugation. Size separates BEVs from bacteria, flagella and cell debris in
stool; and blood cells, high density lipoproteins (HDLs) and soluble proteins in
blood. Density separates BEVs from fibers, protein aggregates and EEVs in stool;
and low-density lipoproteins (LDLs), very-low-density lipoproteins (VLDLs),
chylomicrons, protein aggregates and EEVs in blood. The procedure is label free,
maintains the integrity of BEVs and ensures reproducibility through the use of
automated liquid handlers. Post-separation BEVs are characterized using
orthogonal biochemical endotoxin and Toll-like receptor-based reporter assays in
combination with proteomics, electron microscopy and oparticle tracking
analysis (NTA) to evaluate BEV quality, abundance, structure and molecular
cargo. Separation and characterization of BEVs from body fluids can be done
within 72 h, is compatible with EEV analysis and can be readily adopted by
researchers experienced in basic molecular biology and extracellular vesicle
analysis. We anticipate that this protocol will expand our knowledge on the
biological heterogeneity, molecular cargo and function of BEVs in human body
fluids and steer the development of laboratory research tools and clinical
diagnostic kits. |
What is the effect of the venom of the cone snail, Conus tulipa? | Thus, C. tulipa venom comprised both paralytic (putative ion channel modulating α-, ω-, μ-, δ-) and non-paralytic (conantokins, con-ikot-ikots, conopressins) conotoxins. | Cotokin-T, a 21-amino acid peptide which induces sleep-like symptoms in young
mice was purified from the venom of the fish-hunting cone snail, Conus tulipa.
The amino acid sequence of the peptide was determined and verified by chemical
synthesis. The peptide has 4 residues of the modified amino acid,
gamma-carboxyglutamate (Gla). The sequence of the peptide is:
Gly-Glu-Gla-Gla-Tyr-Gln-Lys-Met-Leu-Gla-Asn-Leu-Arg-Gla-Ala-Glu-Val-Lys-
Lys-Asn-Ala-NH2. Cotokin-T inhibits N-methyl-D-aspartate (NMDA)
receptor-mediated calcium influx in central nervous system neurons. This
observation suggests that like cotokin-G (a homologous Conus peptide with
recently identified NMDA antagonist activity) cotokin-T has NMDA antagonist
activity. A sequence comparison of cotokins-T and -G identifies the 4 Gla
residues and the N-terminal dipeptide sequence as potential key elements for the
biological activity of this peptide. This study investigated the mode of action of cotokin-T, a 21 amino acid
peptide toxin isolated from the venom of the fish-hunting cone snail Conus
tulipa, on excitatory synaptic transmission in rat hippocampal slices using
intracellular recording techniques. Superfusion of cotokin-T (1-500 nM)
specifically and irreversibly decreased the pharmacologically isolated
N-methyl-D-aspartate receptor (NMDA)-mediated excitatory postsynaptic potential
(EPSPNMDA) in a concentration-dependent manner but had no effect on normal
excitatory synaptic transmission (EPSP). The sensitivity of postsynaptic neurons
to NMDA but not to alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid was
also antagonized by cotokin-T pretreatment. In addition, the
cotokin-T-induced depression of EPSPNMDA could be antagonized by prior
treatment of hippocampal slices with either DL-2-amino-5-phosphonovaleate (10
microM) or ifenprodil (20 microM). However, 7-chlorokynurenic acid (1 microM)
had no effect on the action of cotokin-T. These findings indicated that
cotokin-T modulates the NMDA receptor by an interaction with its glutamate
binding site and polyamine recognition site. Subunit non-selective N-methyl-D-aspartate (NMDA) receptor antagonists reduce
injury-induced pain behavior, but generally produce unacceptable side effects.
In this study, we examined the antinociceptive and motor effects of cone snail
venom-derived peptides, cotokins G and T (conG and conT), which are selective
inhibitors of the NR2B or NR2A and NR2B subtypes of the NMDA receptor,
respectively. We tested the effects of conG and conT in models of tissue
(formalin test), nerve injury (partial sciatic nerve ligation) and
inflammation-induced (intraplantar Complete Freund's Adjuvant; CFA) pain in
mice. In the formalin test, intrathecal (i.t.) conG or conT suppressed the
ongoing pain behavior (ED(50) and 95% confidence intervals (CI), 11 (7-19) and
19 (11-33), respectively) at doses that were 17-27 times lower than those
required to impair motor function (accelerating rotarod treadmill test: ED(50)
and 95% CI, 300 (120-730) and 320 (190-540) pmol, respectively). By comparison,
SNX-111, an N-type voltage-sensitive calcium channel antagonist that is also
derived from cone snail venom, produced significant motor impairment at a dose
(3.0 pmol, i.t.) that was only partially efficacious in the formalin test.
Furthermore, conG reversed the allodynia produced by nerve injury, with greater
potency on thermal (ED50 and 95% CI, 24 (10-55) pmol) than on mechanical
allodynia (59 (33-105) pmol). Finally, a single dose of conG (100 pmol, i.t.)
also reduced CFA-evoked thermal and mechanical allodynia. Taken together, these
results demonstrate that cotokins exhibit potent antinociceptive effects in
several models of injury-induced pain. The study supports the notion that drugs
directed against subtypes of the NMDA receptor, by virtue of their reduced
side-effect profile, hold promise as novel therapeutic agents for the control of
pain. Author information:
(1)Department of Biology, University of Utah, Salt Lake City, UT 84112;
Department of Biology, Copenhagen Biocenter, DK-2200 Copenhagen, Denmark;
[email protected] [email protected].
(2)Department of Biology, University of Utah, Salt Lake City, UT 84112;
(3)University of Utah Molecular Medicine Program, University of Utah School of
Medicine, Salt Lake City, UT 84112; Department of Internal Medicine, Division of
Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine,
Salt Lake City, UT 84112;
(4)Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash
University, Parkville, VIC 3052, Australia;
(5)Department of Biochemistry and Molecular Pharmacology, New York University
Langone Medical Center, New York, NY 10016;
(6)Department of Neurobiology and Anatomy, University of Utah, Salt Lake City,
UT 84112;
(7)University of Utah Molecular Medicine Program, University of Utah School of
Medicine, Salt Lake City, UT 84112; Department of Internal Medicine, Division of
Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine,
Salt Lake City, UT 84112; Department of Biochemistry, University of Utah School
of Medicine, Salt Lake City, UT 84112;
(8)Eccles institute of Human Genetics, University of Utah, Salt Lake City, UT
84112; The Utah Science Technology and Research Initiative Center for Genetic
Discovery, University of Utah, Salt Lake City, UT 84112; and.
(9)Eccles institute of Human Genetics, University of Utah, Salt Lake City, UT
84112;
(10)Department of Biochemistry and Molecular Biology, School of Biomedical
Sciences, Monash University, Clayton, VIC 3800, Australia.
(11)Department of Biology, Copenhagen Biocenter, DK-2200 Copenhagen, Denmark;
(12)Department of Biology, University of Utah, Salt Lake City, UT 84112;
[email protected] [email protected]. Plain Language Summary: Insulin is a hormone critical for maintaining healthy
blood sugar levels in humans. When the insulin system becomes faulty, blood
sugar levels become too high, which can lead to diabetes. At the moment, the
only effective treatment for one of the major types of diabetes are daily
insulin injections. However, designing fast-acting insulin drugs has remained a
challenge. Insulin molecules form clusters (so-called hexamers) that first have
to dissolve in the body to activate the insulin receptor, which plays a key role
in regulating the blood sugar levels throughout the body. This can take time and
can therefore delay the blood-sugar control. In 2015, researchers discovered
that the fish-hunting cone snail Conus geographus uses a specific type of
insulin to capture its prey – fish. The cone snail releases insulin into the
surrounding water and then engulfs its victim with its mouth. This induces
dangerously low blood sugar levels in the fish and so makes them an easy target.
Unlike the human version, the snail insulin does not cluster, and despite
structural differences, can bind to the human insulin receptor. Now,
Ahorukomeye, Disotuar et al. – including some of the authors involved in the
previous study – wanted to find out whether other fish-hunting cone snails also
make insulins and if they differed from the one previously discovered in C.
geographus. The insulin molecules were extracted and analyzed, and the results
showed that the three cone snail species had different versions of insulin – but
none of them formed clusters. Ahorukomeye, Disotuar et al. further revealed that
the snail insulins could bind to the human insulin receptors and could also
reverse high blood sugar levels in fish and mouse models of the disease. This
research may help guide future studies looking into developing fast-acting
insulin drugs for diabetic patients. A next step will be to fully understand how
snail insulins can be active at the human receptor without forming clusters.
Cone snails solved this problem millions of years ago and by understanding how
they have done this, researchers are hoping to redesign current diabetic
therapeutics. Since the snail insulins do not form clusters and should act
faster than currently available insulin drugs, they may lead to better or new
diabetes treatments. Cone snails use separately evolved venoms for prey capture and defence. While
most use a harpoon for prey capture, the Gastridium clade that includes the
well-studied Conus geographus and Conus tulipa, have developed a net hunting
strategy to catch fish. This unique feeding behaviour requires secretion of
"nirvana cabal" peptides to dampen the escape response of targeted fish allowing
for their capture directly by mouth. However, the active components of the
nirvana cabal remain poorly defined. In this study, we evaluated the behavioural
effects of likely nirvana cabal peptides on the teleost model, Danio rerio
(zebrafish). Surprisingly, the cotokins (NMDA receptor antagonists) and/or
conopressins (vasopressin receptor agonists and antagonists) found in C.
geographus and C. tulipa venom failed to produce a nirvana cabal-like effect in
zebrafish. In contrast, low concentrations of the non-competitive adrenoceptor
antagonist ρ-TIA found in C. tulipa venom (EC50 = 190 nM) dramatically reduced
the escape response of zebrafish larvae when added directly to aquarium water.
ρ-TIA inhibited the zebrafish α1-adrenoceptor, confirming ρ-TIA has the
potential to reverse the known stimulating effects of norepinephrine on fish
behaviour. ρ-TIA may act alone and not as part of a cabal, since it did not
synergise with conopressins and/or cotokins. This study highlights the
importance of using ecologically relevant animal behaviour models to decipher
the complex neurobiology underlying the prey capture and defensive strategies of
cone snails. |
Which component of the Influenza A Virus affects mRNA transcription termination? | Defective Pol II termination occurs independently of the ability of the viral NS1 protein to interfere with host mRNA processing. Instead, this termination defect is a common effect of diverse cellular stresses and underlies the production of previously reported downstream-of-gene transcripts (DoGs). | Carbocyclic 2',3'-didehydro-2',3'-dideoxyguanosine (carbovir, NSC 614846) is an
anti-retroviral agent that may be useful in the treatment of AIDS. We have
examined the ability of (-)-etiomeric carbovir triphosphate to inhibit human
immunodeficiency virus type 1 (HIV-1) reverse transcriptase (EC 2.7.7.49). A
comparison of inhibition kinetics was made with 3'-azido-2',3'-dideoxythymidine
triphosphate and phosphonoformate. Inhibition of the reverse transcriptase was
evaluated using poly(rA).oligo(dT)12-18, poly(rC).oligo(dG)12-18, or influenza
virion RNA template with a specific oligodeoxynucleotide as primer. (-)-Carbovir
5'-triphosphate was shown to be a potent inhibitor of HIV-1 reverse
transcriptase with an apparent Ki similar to that of
3'-azido-2',3'-dideoxythymidine triphosphate. Chain elongation studies utilizing
an MS2 RNA template showed that (-)-carbovir 5'-triphosphate terminated
transcription at positions identical to those where dideoxy-GTP terminated. This
indicates that (-)-carbovir 5'-monophosphate is incorporated into the newly
synthesized DNA and terminates transcription at that point. We conclude that
(-)-carbovir 5'-triphosphate is a potent inhibitor of the HIV-1 reverse
transcriptase enzyme and that (-)-carbovir most likely inhibits HIV by activity
at the triphosphate level by a combination of direct competition for binding of
the natural deoxynucleoside triphosphates to the reverse transcriptase and chain
termination. Influenza virus gene 8 codes for two nonstructural proteins (NS1 and NS2) which
are translated, respectively, from a colinear and an interrupted mRNA. To
investigate the mechanism of transcription processing by splicing, cloned
full-length NS DNA was inserted into the late region of an SV40 vector. In this
manner, transcription of NS RNA from the recombit initiates and terminates
using SV40 control sequences. RNA mapping and sequence analysis showed that the
RNA transcripts are processed to produce both the spliced and unspliced NS mRNAs
in approximately equal abundance. The junction sequence of the spliced mRNA is
identical to that of NS2 mRNA found in influenza virus-infected cells. In
addition, another mRNA species was found to contain a chimera splice between
SV40 and NS sequences. Both NS1 and NS2 polypeptides are produced in the
recombit-infected cells as predicted from sequence analysis. These studies
establish that during influenza virus infections processing of the NS1 mRNA
transcript undergoes a mechanism of splicing similar to that occurring with
DNA-directed RNA transcription. Viruses have evolved a number of translational control mechanisms to regulate
the levels of expression of viral proteins on polycistronic mRNAs, including
programmed ribosomal frameshifting and stop codon readthrough. More recently,
another unusual mechanism has been described, that of termination-dependent
re-initiation (also known as stop-start). Here, the AUG start codon of a 3' ORF
(open reading frame) is proximal to the termination codon of a uORF (upstream
ORF), and expression of the two ORFs is coupled. For example, segment 7 mRNA of
influenza B is bicistronic, and the stop codon of the M1 ORF and the start codon
of the BM2 ORF overlap in the pentanucleotide UAAUG (stop codon of M1 is shown
in boldface and start codon of BM2 is underlined). This short review aims to
provide some insights into how this translational coupling process is regulated
within different viral systems and to highlight some of the differences in the
mechanism of re-initiation on prokaryotic, eukaryotic and viral mRNAs. Viruses utilize a number of translational control mechanisms to regulate the
relative expression levels of viral proteins on polycistronic mRNAs. One such
mechanism, that of termination-dependent reinitiation, has been described in a
number of both negative- and positive-strand RNA viruses. Dicistronic RNAs which
exhibit termination-reinitiation typically have a start codon of the 3'-ORF
(open reading frame) proximal to the stop codon of the upstream ORF. For
example, the segment 7 RNA of influenza B is dicistronic, and the stop codon of
the M1 ORF and the start codon of the BM2 ORF overlap in the pentanucleotide
UAAUG (the stop codon of M1 is shown in bold and the start codon of BM2 is
underlined). Recent evidence has highlighted the potential importance of
mRNA-rRNA interactions in reinitiation on caliciviral and influenza B viral
RNAs, probably used to tether 40S ribosomal subunits to the RNA after
termination in time for initiation factors to be recruited to the AUG of the
downstream ORF. The present review summarizes how such interactions regulate
reinitiation in an array of RNA viruses, and discusses what is known about
reinitiation in viruses that do not rely on apparent mRNA-rRNA interactions. Influenza virus intimately associates with host RNA polymerase II (Pol II) and
mRNA processing machinery. Here, we use mammalian native elongating transcript
sequencing (mNET-seq) to examine Pol II behavior during viral infection. We show
that influenza virus executes a two-pronged attack on host transcription. First,
viral infection causes decreased Pol II gene occupancy downstream of
transcription start sites. Second, virus-induced cellular stress leads to a
catastrophic failure of Pol II termination at poly(A) sites, with transcription
often continuing for tens of kilobases. Defective Pol II termination occurs
independently of the ability of the viral NS1 protein to interfere with host
mRNA processing. Instead, this termination defect is a common effect of diverse
cellular stresses and underlies the production of previously reported
downstream-of-gene transcripts (DoGs). Our work has implications for
understanding not only host-virus interactions but also fundamental aspects of
mammalian transcription. Author information:
(1)Department of Microbiology, Icahn School of Medicine at Mount Sinai, New
York, NY, USA.
(2)Global Health and Emerging Pathogens Institute, Icahn School of Medicine at
Mount Sinai, New York, NY, USA.
(3)Department of Obstetrics and Gynecology, Stanford University, Stanford, CA,
USA.
(4)Institute for Stem Cell Biology & Regenerative Medicine, Stanford University,
Stanford, CA, USA.
(5)Department of Medicine (Infectious Diseases), Northwestern University
Feinberg School of Medicine, Chicago, IL, USA.
(6)Department of Cellular and Molecular Pharmacology, University of California,
San Francisco, San Francisco, CA, USA.
(7)Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine
at Mount Sinai, New York, NY, USA.
(8)Institute of Molecular and Cell Biology (IMCB), Agency for Science,
Technology and Research (A*STAR), Singapore, Singapore.
(9)Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological
Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of
Medicine at Mount Sinai, New York, NY, USA.
(10)Life Science Research Centre, Faculty of Science, University of Ostrava,
Ostrava, Czech Republic.
(11)Epinova Epigenetics Discovery Performance Unit, Immuno-Inflammation Therapy
Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, UK.
(12)Laboratory of Immune Cell Epigenetics and Signaling, The Rockefeller
University, New York, NY, USA.
(13)Cancer Cell Biology Programme, Centro Nacional de Investigaciones
Oncológicas, CNIO, Madrid, Spain.
(14)Department of Psychiatry, Department of Genetics and Genomic Sciences, Icahn
School of Medicine at Mount Sinai, New York, NY, USA.
(15)Department of Genomics and Multiscale Biology, Icahn School of Medicine at
Mount Sinai, New York, NY, USA.
(16)Department of Medicine, Clinical Immunology, Icahn School of Medicine at
Mount Sinai, New York, NY, USA.
(17)Influenza Division, National Center for Immunization and Respiratory
Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
(18)Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York,
NY, USA.
(19)Division of Hematology and Oncology, Department of Medicine, Icahn School of
Medicine at Mount Sinai, New York, NY, USA.
(20)Department of Pathology, Icahn School of Medicine at Mount Sinai, New York,
NY, USA.
(21)Division of Infectious Diseases, Department of Medicine, Icahn School of
Medicine at Mount Sinai, New York, NY, USA.
(22)Department of Microbiology, Icahn School of Medicine at Mount Sinai, New
York, NY, USA. [email protected].
(23)Global Health and Emerging Pathogens Institute, Icahn School of Medicine at
Mount Sinai, New York, NY, USA. [email protected]. |
Which two genes are predominantly considered by warfarin initial dosing algorithms? | Polymorphisms in CYP2C9 and VKORC1 are taken into consideration by warfarin initial dosing algorithms. | Author information:
(1)Division of Clinical Pharmacology and Toxicology, The Hospital for Sick
Children, Toronto, ON, Canada.
(2)Department of Pediatrics, Yokohama City University, School of Medicine,
Yokohama, Kanagawa, Japan.
(3)Division of Clinical Research Planning, Department of Development Strategy,
Center for Clinical Research and Development, National Center for Child Health
and Development, Tokyo, Japan.
(4)Institute of Cellular Medicine, Newcastle University and Newcastle upon Tyne
Hospitals, NHS Foundation Trust, Newcastle upon Tyne, UK.
(5)Department of Haematology, The Newcastle upon Tyne Hospitals NHS Foundation
Trust, Newcastle upon Tyne, UK.
(6)Division of Pediatric Hematology/Oncology/BMT, Nationwide Children's
Hospital, Columbus, OH, US.
(7)Division of Pediatric Hematology/Oncology, Department of Pediatrics, Monroe
Carell Jr. Children's Hospital at Vanderbilt, Vanderbilt University Medical
Center, Nashville, TN, US.
(8)Assistance Publique Hôpitaux de Paris, Hôpital Necker Enfants Malades, Unité
Médico-Chirurgicale de Cardiologie Congénitale et Pédiatrique, Centre de
référence M3C, Paris, France.
(9)University Paris Descartes, Paris, France.
(10)Université Paris Descartes, Inserm Unité Mixte de Recherche (UMR)-S, Paris,
France.
(11)University Paris Descartes, INSERM UMR 1147, Paris, France.
(12)Influenza and Emerging Respiratory Pathogens, BC Centre for Disease Control,
Vancouver, BC, Canada.
(13)Division of Translational Therapeutics, Department of Pediatrics, University
of British Columbia, Vancouver, BC, Canada.
(14)Department of Medical Sciences, Clinical Pharmacology and Science for Life
Laboratory, Uppsala University, Uppsala, Sweden.
(15)Department of Pediatrics, Graduate School of Medicine and Pharmaceutical
Sciences, University of Toyama, Toyama, Japan.
(16)Graduate School of Medicine and Pharmaceutical Sciences, University of
Toyama, Toyama, Japan.
(17)Department of Cardiology, Kanagawa Children's Medical Center, Yokohama,
Japan.
(18)Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan.
(19)Department of Clinical Pharmacology & Genetics, School of Pharmaceutical
Sciences, University of Shizuoka, Shizuoka, Japan.
(20)Division of Pediatric Hematology/Oncology, The Hospital for Sick Children,
Toronto, ON, Canada.
(21)Division of Clinical Pharmacology and Toxicology, The Hospital for Sick
Children, Toronto, ON, Canada. [email protected]. WHAT IS KNOWN AND OBJECTIVE: Warfarin is an oral anticoagulant which has been
widely used to treat and prevent thromboembolic events. Managing warfarin
therapy requires careful monitoring and dose titration. This randomized
controlled study was designed to assess the effect of genotype-guided warfarin
anticoagulation in Chinese elderly patients with nonvalvular atrial
fibrillation.
METHODS: 507 adults were randomized to receive initial dosing as determined by
an algorithm containing genetic (VKORC1 and CYP2C9) plus clinical information or
only clinical information. The primary endpoint was the time in therapeutic
range (TTR) over 90 days. Secondary end points included haemorrhagic events,
thrombotic events and mortality.
RESULTS: The TTR was significantly different between genetic group and control
group. The average TTR was (70.80 ± 24.39) % in the genotype-guided group as
compared with (53.44 ± 26.73) % in the control group. This represents a
difference of 17.36% (95% CI, 11.82 to 22.89, P < .001). The cumulative
incidence of total haemorrhagic events, minor haemorrhagic events,
gastrointestinal bleeding and intracerebral bleeding events was not
significantly different between two groups (P > .05). Follow-up showed that the
cumulative incidence of ischaemic stroke events occurred in the genetic group
was significantly lower than that in the control group (2.39% vs 6.82%), and the
genetic group had a significant lower risk than control group in cumulative
incidence of ischaemic stroke events [HR 0.22, (95% CI 0.065 to 0.77), P < .05].
WHAT IS NEW AND CONCLUSION: Genotype-guided dosing could improve the average
TTR, and follow-up result showed that genotype-guided therapy resulted in a
significantly lower risk of ischaemic stroke events. Further research is
required to focus on the clinical benefit of genotype-guided dosing. |
Describe MSstatsTMT | MSstatsTMT is a general statistical approach for relative protein quantification in MS- based experiments with TMT labeling. | Tandem mass tag (TMT) is a multiplexing technology widely-used in proteomic
research. It enables relative quantification of proteins from multiple
biological samples in a single MS run with high efficiency and high throughput.
However, experiments often require more biological replicates or conditions than
can be accommodated by a single run, and involve multiple TMT mixtures and
multiple runs. Such larger-scale experiments combine sources of biological and
technical variation in patterns that are complex, unique to TMT-based workflows,
and challenging for the downstream statistical analysis. These patterns cannot
be adequately characterized by statistical methods designed for other
technologies, such as label-free proteomics or transcriptomics. This manuscript
proposes a general statistical approach for relative protein quantification in
MS- based experiments with TMT labeling. It is applicable to experiments with
multiple conditions, multiple biological replicate runs and multiple technical
replicate runs, and unbalanced designs. It is based on a flexible family of
linear mixed-effects models that handle complex patterns of technical artifacts
and missing values. The approach is implemented in MSstatsTMT, a freely
available open-source R/Bioconductor package compatible with data processing
tools such as Proteome Discoverer, MaxQuant, OpenMS, and SpectroMine. Evaluation
on a controlled mixture, simulated datasets, and three biological investigations
with diverse designs demonstrated that MSstatsTMT balanced the sensitivity and
the specificity of detecting differentially abundant proteins, in large-scale
experiments with multiple biological mixtures. |
Is Tranexamic acid effective for intracerebral haemorrhage? | No. According to clinical trial data tranexamic acid does not improve outcomes of patients with intracerebral haemorrhage. | Author information:
(1)Stroke Trials Unit, Division of Clinical Neuroscience, University of
Nottingham, City Hospital Campus, Nottingham, UK; Stroke, Nottingham University
Hospitals NHS Trust, City Hospital Campus, Nottingham, UK. Electronic address:
[email protected].
(2)Stroke Trials Unit, Division of Clinical Neuroscience, University of
Nottingham, City Hospital Campus, Nottingham, UK.
(3)Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
(4)Department of Neurology, Semmelweis University, Budapest, Hungary.
(5)The First University Clinic of Tbilisi State Medical University, Tbilisi,
Georgia.
(6)Department of Neurology, Bispebjerg and Frederiksberg Hospital, University of
Copenhagen, Copenhagen, Denmark.
(7)Neurology Unit, Azienda Socio Sanitaria Territoriale di Mantova, Mantua,
Italy.
(8)Stroke Service, Adelaide and Meath Hospital, Tallaght, Ireland.
(9)2nd Department of Neurology, Institute of Psychiatry and Neurology, Warsaw,
Poland.
(10)Radiological Sciences, Division of Clinical Neuroscience, University of
Nottingham, Queens Medical Centre Campus, Nottingham, UK; NIHR Nottingham
Biomedical Research Centre, Nottingham, UK.
(11)Nottingham Clinical Trials Unit, University of Nottingham, Queen's Medical
Centre, Nottingham, UK.
(12)UGC de Medicina Intensiva, Hospital Universitario Virgen del Rocío,
Instituto de Biomedicina de Sevilla, Consejo Superior de Investigaciones
Científicas, Universidad de Sevilla, Seville, Spain.
(13)Vascular Medicine, Division of Medical Sciences and Graduate Entry Medicine,
University of Nottingham, Royal Derby Hospital Centre, Derby, UK.
(14)Stroke Trials Unit, Division of Clinical Neuroscience, University of
Nottingham, City Hospital Campus, Nottingham, UK; Stroke, Nottingham University
Hospitals NHS Trust, City Hospital Campus, Nottingham, UK.
(15)Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet,
Stockholm, Sweden.
(16)Stroke Trials Unit, Division of Clinical Neuroscience, University of
Nottingham, City Hospital Campus, Nottingham, UK; Stroke, Nottingham University
Hospitals NHS Trust, City Hospital Campus, Nottingham, UK; Department of
Medicine, National University of Malaysia, Kuala Lumpur, Malaysia.
(17)Department of Neurology, Selcuk University Medical Faculty, Konya, Turkey.
(18)Department of Medical Statistics, London School of Hygiene & Tropical
Medicine, London, UK.
(19)Clinical Trials Unit, London School of Hygiene & Tropical Medicine, London,
UK.
(20)Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research
Centre, University of Leicester, Leicester, UK.
(21)Stroke Research, Faculty of Medicine and Health Sciences, Keele University,
Staffordshire, UK.
(22)Stroke Center, Neurology and Department of Clinical Research, University
Hospital, University Basel, Basel, Switzerland.
(23)Division of Neurosurgery, Department of Surgery, National University of
Malaysia, Kuala Lumpur, Malaysia.
(24)Stroke Research Centre, UCL Institute of Neurology and National Hospital for
Neurology and Neurosurgery, University College London, London, UK.
(25)School of Economics, University of Nottingham, University Park, Nottingham,
UK. PURPOSE: Haematoma expansion is a devastating complication of intracerebral
haemorrhage (ICH) with no established treatment. Tranexamic acid had been an
effective haemostatic agent in reducing post-operative and traumatic bleeding.
We review current evidence examining the efficacy of tranexamic acid in
improving clinical outcome after ICH.
METHOD: We searched MEDLINE, EMBASE, CENTRAL and clinical trial registers for
studies using search strategies incorporating the terms 'intracerebral
haemorrhage', 'tranexamic acid' and 'antifibrinolytic'. Authors of ongoing
clinical trials were contacted for further details.
FINDINGS: We screened 268 publications and retrieved 17 articles after
screening. Unpublished information from three ongoing clinical trials was
obtained. We found five completed studies. Of these, two randomised controlled
trials (RCTs) comparing intravenous tranexamic acid to placebo (n = 54) reported
no significant difference in death or dependency. Three observational studies
(n = 281) suggested less haematoma growth with rapid tranexamic acid infusion.
There are six ongoing RCTs (n = 3089) with different clinical exclusions,
imaging selection criteria (spot sign and haematoma volume), time window for
recruitment and dosing of tranexamic acid.
DISCUSSION: Despite their heterogeneity, the ongoing trials will provide key
evidence on the effects of tranexamic acid on ICH. There are uncertainties of
whether patients with negative spot sign, large haematoma, intraventricular
haemorrhage, or poor Glasgow Coma Scale should be recruited. The time window for
optimal effect of haemostatic therapy in ICH is yet to be established.
CONCLUSION: Tranexamic acid is a promising haemostatic agent for ICH. We await
the results of the trials before definite conclusions can be drawn. INTRODUCTION: Seizures are common after intracerebral haemorrhage. Tranexamic
acid increases the risk of seizures in non-intracerebral haemorrhage population
but its effect on post-intracerebral haemorrhage seizures is unknown. We
explored the risk factors and outcomes of seizures after intracerebral
haemorrhage and if tranexamic acid increased the risk of seizures in the
Tranexamic acid for IntraCerebral Haemorrhage-2 trial.
PATIENTS AND METHODS: Seizures were reported prospectively up to day 90. Cox
regression analyses were used to determine the predictors of seizures within
90 days and early seizures (≤7 days). We explored the effect of early seizures
on day 90 outcomes.
RESULTS: Of 2325 patients recruited, 193 (8.3%) had seizures including 163
(84.5%) early seizures and 30 (15.5%) late seizures (>7 days). Younger age
(adjusted hazard ratio (aHR) 0.98 per year increase, 95% confidence interval
(CI) 0.97-0.99; p = 0.008), lobar haematoma (aHR 5.84, 95%CI 3.58-9.52;
p < 0.001), higher National Institute of Health Stroke Scale (aHR 1.03, 95%CI
1.01-1.06; p = 0.014) and previous stroke (aHR 1.66, 95%CI 1.11-2.47; p = 0.013)
were associated with early seizures. Tranexamic acid did not increase the risk
of seizure within 90 days. Early seizures were associated with worse modified
Rankin Scale (adjusted odds ratio (aOR) 1.79, 95%CI 1.12-2.86, p = 0.015) and
increased risk of death (aOR 3.26, 95%CI 1.98-5.39; p < 0.001) at day
90.Discussion and conclusion: Lobar haematoma was the strongest independent
predictor of early seizures after intracerebral haemorrhage. Tranexamic acid did
not increase the risk of post-intracerebral haemorrhage seizures in the first
90 days. Early seizures resulted in worse functional outcome and increased risk
of death. |
Are interferons defensive proteins? | Yes,
The innate immune system, in particular the type I interferon (IFN) response, is a powerful defence against virus infections. | The interferon-induced GTP-binding protein Mx is responsible for a specific
antiviral state against a broad spectrum of viral infections that are induced by
type-I interferons (IFN α/β) in different vertebrates. In this study, the Mx
gene was isolated from the constructed mullet cDNA database. Structural features
of mullet Mx (MuMx) were analyzed using different in-silico tools. The pairwise
comparison revealed that the MuMx sequence was related to Stegastes partitus Mx
with an 83.7% sequence identity, whereas MuMx was clustered into the teleost
category in the phylogentic analysis. Sequence alignment showed that the
dynamin-type guanine nucleotide-binding domain (G_DYNAMIN_2), central
interactive domain (CID), and GTPase effector domain (GED) were conserved among
Mx counterparts. The transcriptional expression of MuMx was the highest in blood
cells from unchallenged fish. The temporal mRNA profile showed that MuMx
expression was significantly elevated in all tissues, including blood, spleen,
head kidney, liver, and gills after the injection of polyinosinic-polycytidylic
acid (poly I:C) at many time points. Moreover, MuMx expression increased
slightly, in the blood, spleen, and head kidney at a few time points after the
injection of lipopolysaccharide (LPS) and Lactococcus garvieae (L. garvieae).
Results of the subcellular localization analysis confirmed that the MuMx protein
was highly expressed in the cytoplasm. The analysis of the gene expression of
the viral hemorrhagic septicemia virus (VHSV) under conditions of MuMx
overexpression confirmed the significant inhibition of viral transcripts. The
cell viability (MTT) assay and VHSV titer quantification with the presence of
MuMx indicated a significant reduction in virus replication. Collectively, these
findings suggest that Mx is a specific immune-related gene that elicits crucial
antiviral functions against viral antigens in the mullet fish. In response to viral infections, various pattern recognition receptors (PRRs)
are activated for the production of type I interferon (IFN I). As a center of
these receptor responses, TANK binding kinase-1 (TBK1) activates interferon
regulatory factor 3 (IRF3). SRC is a member of Src family kinases (SFK) which
participates in TBK1-mediated IFN I signaling pathway. In mammals, the
immunological function of SRC is depended on its interaction with TBK1. To date,
SRC has not been studied in fish. In this paper, we cloned the ORF of grass carp
(Ctenopharyngodon idellus) SRC (CiSRC). CiSRC has a closer relationship with
Sinocyclocheilus rhinocerous SRC (SrSRC). The expression level of CiSRC was
significantly up-regulated following poly (I:C) stimulation in grass carp
tissues and cells. Subcellular localization results showed that CiSRC is located
both in the cytoplasm and nucleus, while CiTBK1 is only located in the cytoplasm
of CIK cells. When GFP-CiSRC and FLAG-CiTBK1 were co-transfected into CIK cells,
we found that they were co-localized in the cytoplasm. GST-pulldown and
Co-immunoprecipitation analysis revealed that CiSRC and CiSRC tyrosine kinase
domain deletion mutant (SRC-ΔTyrkc) can interact with CiTBK1, respectively.
CiSRC promotes the phosphorylation of CiTBK1. Furthermore, the phosphorylation
of TBK1 is more strongly under poly (I:C) stimulation. We also demonstrated that
SRC can up-regulate IFN I expression. These results above unraveled that CiSRC
initiates innate immune response by binding to and then up-regulating the
phosphorylation of TBK1. |
What effect does Methylsulfonylmethane (MSM) have on inflammation? | Methylsulfonylmethane (MSM) is a sulfur-based nutritional supplement that is purported to have pain and inflammation-reducing effects. | Methylsulfonylmethane (MSM) is a popular dietary supplement used in a variety of
conditions including pain, inflammation, allergies, arthritis, parasitic
infections and the maintece of normal keratin levels in hair, skin and nails.
Despite its popularity, there is little published toxicology data on MSM. The
objective of this study was to evaluate the acute and subchronic toxicity of MSM
in rats at a dose five to seven times the maximum recommended dose in humans.
MSM administered in a single gavage dose of 2 g/kg resulted in no adverse events
or mortality. MSM administered as a daily dose of 1.5 g/kg for 90 days by gavage
resulted in no adverse events or mortality. Necropsy did not reveal any gross
pathological lesions or changes in organ weights. Renal histology of treated
animals was normal. It is concluded that MSM is well tolerated in rats at an
acute dose of 2 g/kg and at a subacute chronic dose of 1.5 g/kg. OBJECTIVE: Glucosamine, classified as a slow-acting drug in osteoarthritis
(SADOA), is an efficacious chondroprotective agent. Methylsulfonylmethane (MSM),
the isoxidised form of dimethyl-sulfoxide (DSMO), is an effective natural
analgesic and anti-inflammatory agent. The aim of this study was to compare the
efficacy and safety of oral glucosamine (Glu), methylsulfonylmethane (MSM),
their combination and placebo in osteoarthritis of the knee.
PATIENTS AND DESIGN: A total of 118 patients of either sex with mild to moderate
osteoarthritis were included in the study and randomised to receive either Glu
500mg, MSM 500mg, Glu and MSM or placebo capsules three times daily for 12
weeks. Patients were evaluated at 0 (before drug administration), 2, 4, 8 and 12
weeks post-treatment for efficacy and safety. The efficacy parameters studied
were the pain index, the swelling index, visual analogue scale pain intensity,
15m walking time, the Lequesne index, and consumption of rescue medicine.
RESULTS: Glu, MSM and their combination significantly improved signs and
symptoms of osteoarthritis compared with placebo. There was a statistically
significant decrease in mean (+/- SD) pain index from 1.74 +/- 0.47 at baseline
to 0.65 +/- 0.71 at week 12 with Glu (p < 0.001). MSM significantly decreased
the mean pain index from 1.53 +/- 0.51 to 0.74 +/- 0.65, and combination
treatment resulted in a more significant decrease in the mean pain index (1.7
+/- 0.47 to 0.36 +/- 0.33; p < 0.001). After 12 weeks, the mean swelling index
significantly decreased with Glu and MSM, while the decrease in swelling index
with combination therapy was greater (1.43 +/- 0.63 to 0.14 +/- 0.35; p < 0.05)
after 12 weeks. The combination produced a statistically significant decrease in
the Lequesne index. All treatments were well tolerated.
CONCLUSION: Glu, MSM and their combination produced an analgesic and
anti-inflammatory effect in osteoarthritis. Combination therapy showed better
efficacy in reducing pain and swelling and in improving the functional ability
of joints than the individual agents. All the treatments were well tolerated.
The onset of analgesic and anti-inflammatory activity was found to be more rapid
with the combination than with Glu. It can be concluded that the combination of
MSM with Glu provides better and more rapid improvement in patients with
osteoarthritis. Methylsulfonylmethane (MSM), also known as dimethyl sulfone and methyl sulfone,
is an organic sulfur-containing compound that occurs naturally in a variety of
fruits, vegetables, grains, and animals, including humans. In the present study,
we demonstrated the anti-inflammatory effects of MSM in lipopolysaccharide
(LPS)-stimulated murine macrophages, RAW264.7 cells. MSM significantly inhibited
the release of nitric oxide and prostaglandin E(2) by alleviating the expression
of inducible nitric oxide synthase and cyclooxygenase-2 in LPS-stimulated
RAW264.7 cells. Furthermore, the levels of interleukin-6 and tumor necrosis
factor-alpha were decreased by MSM treatment in cell culture supernatants.
Further study indicated that the translocation of the p65 subunit of nuclear
factor (NF)-kappaB to the nucleus was inhibited by MSM treatment in
LPS-stimulated RAW264.7 cells, in which it helped block degradation of inhibitor
of NF-kappaB. In addition, in vivo studies demonstrated that topical
administration of MSM at 500-1250 microg/ear resulted in similar inhibitory
activities in 12-O-tetradecanoylphorbol 13-acetate-induced mouse ear edema.
Collectively, theses results indicate that MSM inhibits LPS-induced release of
pro-inflammatory mediators in murine macrophages through downregulation of
NF-kappaB signaling. Methylsulfonylmethane (MSM), which is one of the popular ingredients of
so-called health foods in Japan, is expected to relieve inflammation in
arthritis and allergies. However, there is no scientific evidence to confirm the
efficacy and safety of MSM in detail. In this study, we examined the effects of
MSM on cartilage formation in growing rats (G) and cartilage degradation in
STR/Ort mice (A), an accepted human osteoarthritis (OA) model. For cartilage
formation study, 6-week-old growing male Wister rats were assigned to four
groups to receive a control or MSM-containing diet. To examine the efficacy of
MSM on the cartilage of OA model mouse, 10-week-old male STR/OrtCrlj mice were
assigned to three groups to receive a control or MSM-containing diet. The
dosages used were amounts equal to the recommended supplements for humans
[0.06 g/kg body weight (BW)/day: MSM1G and MSM1A], 10 fold higher (0.6 g/kg
BW/day: MSM10G and MSM10A), and 100 fold higher (6 g/kg BW/day: MSM100G). Intake
of MSM for 4 weeks did not affect cartilage formation in the knee joint in
growing rats. Body, liver, and spleen weight in the MSM100G group were
significantly lower than those in the control group. Intake of MSM for 13 weeks
decreased degeneration of the cartilage at the joint surface in the knee joints
in STR/Ort mice in a dose-dependent manner. These results suggest that
appropriate intake of MSM is possibly effective in OA model mice; however,
intake of large amounts of MSM induced atrophy of several organs. BACKGROUND: Methylsulfonylmethane (MSM) has been reported to provide
anti-inflammatory and antioxidant effects in both animal and man. Strenuous
resistance exercise has the potential to induce both inflammation and oxidative
stress. Using a pilot (proof of concept) study design, we determined the
influence of MSM on markers of exercise recovery and performance in healthy men.
METHODS: Eight, healthy men (27.1 ± 6.9 yrs old) who were considered to be
moderately exercise-trained (exercising <150 minutes per week) were randomly
assigned to ingest MSM at either 1.5 grams per day or 3.0 grams per day for 30
days (28 days before and 2 days following exercise). Before and after the 28 day
intervention period, subjects performed 18 sets of knee extension exercise in an
attempt to induce muscle damage (and to be used partly as a measure of exercise
performance). Sets 1-15 were performed at a predetermined weight for 10
repetitions each, while sets 16-18 were performed to muscular failure. Muscle
soreness (using a 5-point Likert scale), fatigue (using the fatigue-inertia
subset of the Profile of Mood States), blood antioxidant status (glutathione and
Trolox Equivalent Antioxidant Capacity [TEAC]), and blood homocysteine were
measured before and after exercise, pre and post intervention. Exercise
performance (total work performed during sets 16-18 of knee extension testing)
was also measured pre and post intervention.
RESULTS: Muscle soreness increased following exercise and a trend was noted for
a reduction in muscle soreness with 3.0 grams versus 1.5 grams of MSM
(p = 0.080), with a 1.0 point difference between dosages. Fatigue was slightly
reduced with MSM (p = 0.073 with 3.0 grams; p = 0.087 for both dosages
combined). TEAC increased significantly following exercise with 3.0 grams of MSM
(p = 0.035), while homocysteine decreased following exercise for both dosages
combined (p = 0.007). No significant effects were noted for glutathione or total
work performed during knee extension testing (p > 0.05).
CONCLUSION: MSM, especially when provided at 3.0 grams per day, may favorably
influence selected markers of exercise recovery. More work is needed to extend
these findings, in particular using a larger sample of subjects and the
inclusion of additional markers of exercise recovery and performance. Methylsulfonylmethane (MSM) is a natural organosulfur compound that exhibits
antioxidative and anti-inflammatory effects. This study was carried out to
investigate the effect of MSM on paraquat (PQ)-induced acute lung and liver
injury in mice. A single dose of PQ (50 mg/kg, i.p.) induced acute lung and
liver toxicity. Mice were treated with MSM (500 mg/kg/day, i.p.) for 5 days. At
the end of the experiment, animals were euthanized, and lung and liver tissues
were collected for histological and biochemical analysis. Tissue samples were
used to determine malondialdehyde (MDA), myeloperoxidase (MPO), catalase (CAT),
superoxide dismutase (SOD), glutathione (GSH), and tumor necrosis factor-α
(TNF-α) levels. Blood samples were used to measure plasma alanine transaminase
(ALT), γ-glutamyl transferase (GGT), and alkaline phosphatase (ALP).
Histological examination indicated that MSM decreased lung and liver damage
caused by PQ. Biochemical results showed that MSM treatment significantly
reduced tissue levels of MDA, MPO, and TNF-α, while increased the levels of SOD,
CAT, and GSH compared with PQ group. MSM treatment also significantly reduced
plasma levels of ALT, GGT, and ALP. These findings suggest that MSM as a natural
product attenuates PQ-induced pulmonary and hepatic oxidative injury. Methylsulfonylmethane (MSM) is an organosulfur compound and the health benefits
associated with MSM include inflammation. Although MSM has been shown to have
various physiological effects, no study has yet focused on inflammasome
activation. The inflammasome is a multiprotein complex that serves as a platform
for caspase 1-dependent proteolytic maturation and secretion of interleukin-1β
(IL-1β). In this study, we tested the effect of MSM on inflammasome activation
using mouse and human macrophages. In our results, MSM significantly attenuated
NLRP3 inflammasome activation in lipopolysaccharide-primed macrophages, although
it had no effect on NLCR4 or AIM2 inflammasome activation. Extracts of
MSM-enriched vegetables presented the same inhibitory effect on NLRP3
inflammasome activation as MSM. MSM also attenuated the transcriptional
expression of IL-1α, IL-1β, IL-6, and NLRP3. Taken together, these results show
that MSM has anti-inflammatory characteristics, interrupts NLRP3 inflammasome
activation, and inhibits pro-cytokine expression. We further confirmed the
intracellular mechanism of MSM in relation to NLRP3 inflammasome activation,
followed by comparison with that of DMSO. Both chemicals showed a synergic
effect on anti-NLRP3 activation and attenuated production of mitochondrial
reactive oxygen species (ROS). Thus, MSM is a selective inhibitor of NLRP3
inflammasome activation and can be developed as a supplement to control several
metabolic disorders. Until now glucosamine sulfate (GS) has been the most widely used supplement and
has been shown to be efficacious in the treatment of osteoarthritis (OA).
Methylsulfonylmethane (MSM) and boswellic acids (BA) are new effective
supplements for the management of inflammation and joint degeneration, according
to previous experimental studies. The aim of our study is to test the
effectiveness of association of MSM and BA in comparison with GS in knee
arthritis.In this prospective randomized clinical trial, MEBAGA
(Methylsulfonylmethane and Boswellic Acids versus Glucosamine sulfate in the
treatment of knee Arthritis), 120 participants affected by arthritis of the knee
were randomly assigned to an experimental group (MB group) or a control group
(GS group) treated for 60 days with 5 g of MSM and 7.2 mg of BA or with 1500 mg
of GS daily, respectively. At the 2-month (T1) and 6-months (T2) follow-up , the
efficacy of these two nutraceuticals was assessed using the visual analog pain
scale (VAS) and the Lequesne Index (LI) for joint function, along with the use
of anti-inflammatory drugs (non-steroidal anti-inflammatory drugs and
anti-cyclooxygenase-2).The repeated measures ANOVA analysis shows that for VAS,
LI, and the use of anti-inflammatory drugs scores there are improvements due to
the time in the two groups (respectively, F=26.0; P<0.0001; F=4.15; P=0.02;
F=3.38; P=0.04), with a tendency to better values for the MB group at T2.On the
basis of these preliminary data, we could support the efficacy of the MSM in
association with BA in the treatment of OA. These results are consistent with
the anti-inflammatory and chondroprotective effects previously occurred in
experimental studies. This new combination of integration (MSM and BS) has
presented good results and satisfactory in comparison with GS, until now the
cornerstone of the treatment of arthritis in according to guidelines. Osteoclast differentiation is dependent on the activities of receptor activator
NF-kB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF). Given
that RANKL plays a critical role in osteoclast formation and bone resorption,
any new compounds found to alter its activity would be predicted to have
therapeutic potential for disorders associated with bone loss.
Methylsulfonylmethane (MSM) is a naturally occurring sulfur compound with
well-documented anti-oxidant and anti-inflammatory properties; currently its
effects on osteoclast differentiation are unknown. We sought to investigate
whether MSM could regulate osteoclastogenesis, and if so, its mechanism of
action. In this study, we investigated the effects of MSM on RANKL-induced
osteoclast differentiation, together with STAT3's involvement in the expression
of osteoclastic gene markers. These experiments were conducted using bone marrow
derived macrophages (BMMs) and cell line material, together with analyses that
interrogated both protein and mRNA levels, as well as signaling pathway
activity. Although MSM was not toxic to osteoclast precursors, MSM markedly
inhibited RANKL-induced TRAP activity, multinucleated osteoclast formation, and
bone resorptive activity. Additionally, the expression of several
osteoclastogenesis-related marker genes, including TRAF6, c-Fos, NFATc1,
cathepsin K, and OSCAR were suppressed by MSM. MSM mediated suppression of
RANKL-induced osteoclastogenesis involved inhibition of ITAM signaling effectors
such as PLCγ and Syk, with a blockade of NF-kB rather than MAPK activity.
Furthermore, MSM inhibited RANKL-induced phosphorylation of STAT3 Ser727.
Knockdown of STAT3 using shRNAs resulted in reduced RANKL-mediated
phosphorylation of Ser727 STAT3, and TRAF6 in cells for which depletion of STAT3
was confirmed. Additionally, the expression of RANKL-induced osteoclastogenic
marker genes were significantly decreased by MSM and STAT3 knockdown. Taken
together, these results indicate that STAT3 plays a pivotal role in
RANKL-induced osteoclast formation, and that MSM can attenuate RANKL-induced
osteoclastogenesis by blocking both NF-kB and STAT3 activity. Background. Inflammation is associated with strenuous exercise and
methylsulfonylmethane (MSM) has been shown to have anti-inflammatory properties.
Methods. Physically active men were supplemented with either placebo or MSM (3
grams per day) for 28 days before performing 100 repetitions of eccentric knee
extension exercise. Ex vivo and in vitro testing consisted of evaluating
cytokine production in blood (whole blood and isolated peripheral blood
mononuclear cells (PBMCs)) exposed to lipopolysaccharide (LPS), before and
through 72 hours after exercise, while in vivo testing included the evaluation
of cytokines before and through 72 hours after exercise. Results. LPS
stimulation of whole blood after MSM supplementation resulted in decreased
induction of IL-1β, with no effect on IL-6, TNF-α, or IL-8. After exercise,
there was a reduced response to LPS in the placebo, but MSM resulted in robust
release of IL-6 and TNF-α. A small decrease in resting levels of proinflammatory
cytokines was noted with MSM, while an acute postexercise increase in IL-10 was
observed with MSM. Conclusion. Strenuous exercise causes a robust inflammatory
reaction that precludes the cells from efficiently responding to additional
stimuli. MSM appears to dampen the release of inflammatory molecules in response
to exercise, resulting in a less incendiary environment, allowing cells to still
have the capacity to mount an appropriate response to an additional stimulus
after exercise. BACKGROUND: Oxidative stress and muscle damage occur during exhaustive bouts of
exercise, and many runners report pain and soreness as major influences on
changes or breaks in training regimens, creating a barrier to training
persistence. Methylsulfonylmethane (MSM) is a sulfur-based nutritional
supplement that is purported to have pain and inflammation-reducing effects. To
investigate the effects of MSM in attenuating damage associated with physical
exertion, this randomized, double-blind, placebo-controlled study evaluated the
effects of MSM supplementation on exercise-induced pain, oxidative stress and
muscle damage.
METHODS: Twenty-two healthy females (n = 17) and males (n = 5) (age
33.7 ± 6.9 yrs.) were recruited from the 2014 Portland Half-Marathon registrant
pool. Participants were randomized to take either MSM (OptiMSM®) (n = 11), or a
placebo (n = 11) at 3 g/day for 21 days prior to the race and for two days after
(23 total). Participants provided blood samples for measurement of markers of
oxidative stress, and completed VAS surveys for pain approximately one month
prior to the race (T0), and at 15 min (T1), 90 min (T2), 1 Day (T3), and 2 days
(T4) after race finish. The primary outcome measure 8-hydroxy-2-deoxyguanine
(8-OHdG) measured oxidative stress. Secondary outcomes included malondialdehyde
(MDA) for oxidative stress, creatine kinase (CK) and lactate dehydrogenase (LDH)
as measures of muscle damage, and muscle (MP) and joint pain (JP) recorded using
a 100 mm Visual Analogue Scale (VAS). Data were analyzed using repeated and
multivariate ANOVAs, and simple contrasts compared post-race time points to
baseline, presented as mean (SD) or mean change (95% CI) where appropriate.
RESULTS: Running a half-marathon induced significant increases in all outcome
measures (p < 0.001). From baseline, 8-OHdG increased significantly at T1 by
1.53 ng/mL (0.86-2.20 ng/mL CI, p < 0.001) and T2 by 1.19 ng/mL (0.37-2.01 ng/mL
CI, p < 0.01), and fell below baseline levels at T3 by -0.46 ng/mL (-1.18-0.26
CI, p > 0.05) and T4 by -0.57 ng/mL (-1.27-0.13 CI, p > 0.05). MDA increased
significantly at T1 by 7.3 μM (3.9-10.7 CI, p < 0.001). Muscle damage markers CK
and LDH saw significant increases from baseline at all time-points (p < 0.01).
Muscle and joint pain increased significantly from baseline at T1, T2, and T3
(p < 0.01) and returned to baseline levels at T4. Time-by-treatment results did
not reach statistical significance for any outcome measure, however, the MSM
group saw clinically significant (Δ > 10 mm) reductions in both muscle and joint
pain.
CONCLUSION: Participation in a half-marathon was associated with increased
markers of oxidative stress, muscle damage, and pain. MSM supplementation was
not associated with a decrease from pre-training levels of oxidative stress or
muscle damage associated with an acute bout of exercise. MSM supplementation
attenuated post-exercise muscle and joint pain at clinically, but not
statistically significant levels. The development of various cardiovascular diseases (CVDs) are associated with
chronic inflammation. Tumor necrosis factor α (TNF-α) is a pro-inflammatory
cytokine that activates the nuclear factor-κB (NF-κB) signaling pathway, leading
to increased inflammatory cytokine expression, such as interleukin-6 (IL-6).
Interventions to reduce each of these factors have been demonstrated to reduce
the development of CVD. Methylsulfonylmethane (MSM) is a naturally occurring
compound that demonstrates anti-inflammatory effects in humans and various
animal and cell culture models. The effects of MSM include decreased NF-κB
activation, decreased expression of TNF-α, and IL-6. However, the effects of MSM
within the heart have not yet been examined. Therefore, the purpose of this
investigation was to determine whether MSM protects cardiac cells from
inflammation that occurs in response to pro-inflammatory stimuli. A novel
immortalized human ventricular cardiomyocyte cell line, designated Ac16,
developed and characterized in the laboratory of Dr. Mercy Davidson, Columbia
Invention Report No. 823, U.S. patent No. 7,223,599 were utilized. Cells were
treated with TNF-α, alone or in combination with MSM. To confirm an appropriate
dosage of MSM, the effect of various concentrations on cell viability, and IL-6
production were examined. The effect of MSM on transcript expression of
pro-inflammatory markers and activation of NF-κB were examined with the
established dose by real-time quantitative PCR and western blot, respectively.
MSM treatment combined with TNF-α significantly decreased IL-6 production and
transcript expression compared to TNF-α alone. These findings indicate that MSM
may protect against inflammation in the heart, and thereby protect against
inflammation-linked CVDs. Further study is warranted to determine the effect of
MSM on cardiovascular health outcomes. High glucose-induced inflammation leads to atherosclerosis, which is considered
a major cause of death in type 1 and type 2 diabetic patients. Nuclear
factor-kappa B (NF-κB) plays a central role in high glucose-induced inflammation
and is activated through toll-like receptors (TLRs) as well as canonical and
protein kinase C-dependent (PKC) pathways. Non-toxic sulfur (NTS) and
methylsulfonylmethane (MSM) are two sulfur-containing natural compounds that can
induce anti-inflammation. Using Western blotting, real-time polymerase chain
reaction, and flow cytometry, we found that high glucose-induced inflammation
occurs through activation of TLRs. An effect of NTS and MSM on canonical and
PKC-dependent NF-κB pathways was also demonstrated by western blotting. The
effects of proinflammatory cytokines were investigated using a chromatin
immunoprecipitation assay and enzyme-linked immunosorbent assay. Our results
showed inhibition of the glucose-induced expression of TLR2 and TLR4 by NTS and
MSM. These sulfur compounds also inhibited NF-κB activity through reactive
oxygen species (ROS)-mediated canonical and PKC-dependent pathways. Finally, NTS
and MSM inhibited the high glucose-induced expression of interleukin (IL)-1β,
IL-6, and tumor necrosis factor-α and binding of NF-κB protein to the DNA of
proinflammatory cytokines. Together, these results suggest that NTS and MSM may
be potential drug candidates for anti-inflammation therapy. BACKGROUND: Methylsulfonylmethane (MSM) is a commonly used diet supplement
believed to decrease the inflammation in joints and fastens recovery in
osteoarthritis, gastric mucosal injury, or obesity-related disorders. It was
also suggested that MSM might play a beneficial role in cancer treatment.
PURPOSE: So far, the MSM might have a potentially beneficial effect in
endometrial cancer (EC) treatment.
STUDY DESIGN: This study evaluated the effect and usefulness of MSM in
combinatory therapy with known drug doxorubicin (DOX).
METHODS: The effect of combinational treatment of MSM and DOX on the induction
of apoptosis was evaluated in EC cell lines (ISHIKAWA, MFE-296, MFE-280).
RESULTS: We observed that MSM itself induces apoptosis in EC cell lines, and
pre-treatment with MSM for 24 h increases the sensitivity of EC cells to
DOX-induced apoptosis and DNA damage and that effect might be regulated by
p42/44 (Erk1/2) MAPK and Akt (protein kinase B).
CONCLUSION: These results for the first time show that MSM might act as a
sensitizer of EC cells to known drugs, for which EC cells quickly acquire
resistance. Graphical abstract. Rotator cuff tears (RCTs) and rotator cuff disease (RCD) are important causes of
disability in middle-aged individuals affected by nontraumatic shoulder
dysfunctions. Our previous studies have demonstrated that four different
hyaluronic acid preparations (HAPs), including Artrosulfur® hyaluronic acid (HA)
(Alfakjn S.r.l., Garlasco, Italy), may exert a protective effect in human
RCT-derived tendon cells undergoing oxidative stress damage. Recently,
methylsulfonylmethane (MSM) (Barentz, Paderno Dugo, Italy) has proven to have
anti-inflammatory properties and to cause pain relief in patients affected by
tendinopathies. This study aims at evaluating three preparations (Artrosulfur®
HA, MSM, and Artrosulfur® MSM + HA) in the recovery from hydrogen
peroxide-induced oxidative stress damage in human tenocyte. Cell proliferation,
Lactate Dehydrogenase (LDH) release, and inducible nitric oxide synthases (iNOS)
and prostaglandin E2 (PGE2) modulation were investigated. In parallel,
expression of metalloproteinases 2 (MMP2) and 14 (MMP14) and collagen types I
and III were also examined. Results demonstrate that Artrosulfur® MSM + HA
improves cell escape from oxidative stress by decreasing cytotoxicity and by
reducing iNOS and PGE2 secretion. Furthermore, it differentially modulates MMP2
and MMP14 levels and enhances collagen III expression after 24 h, proteins
globally related to rapid acceleration of the extracellular matrix (ECM)
remodelling and thus tendon healing. By improving the anti-cytotoxic effect of
HA, the supplementation of MSM may represent a feasible strategy to ameliorate
cuff tendinopathies. |
What is particular about the 3D structure of the inactive X chromosome? | The mammalian inactive X chromosome (Xi) condenses into a bipartite structure with two superdomains of frequent long-range contacts, separated by a hinge region. This specific bipartite organization of the inactive X chromosome that probably plays an important role in maintenance of gene silencing. | The 3D folding structure formed by different genomic regions of a chromosome is
still poorly understood. So far, only relatively simple geometric features, like
distances and angles between different genomic regions, have been evaluated.
This work is concerned with more complex geometric properties, i.e., the
complete shape formed by genomic regions. Our work is based on statistical shape
theory and we use different approaches to analyze the considered structures,
e.g., shape uniformity test, 3D point-based registration, Fisher distribution,
and 3D non-rigid image registration for shape normalization. We have applied
these approaches to analyze 3D microscopy images of the X-chromosome where four
consecutive genomic regions (BACs) have been simultaneously labeled by
multicolor FISH. We have acquired two sets of four consecutive genomic regions
with an overlap of three regions. From the experimental results, it turned out
that for all data sets the complete structure is non-random. In addition, we
found that the shapes of active and inactive X-chromosomal genomic regions are
statistically independent. Moreover, we reconstructed the average 3D structure
of chromatin in a small genomic region (below 4 Mb) based on five BACs resulting
from two overlapping four BAC regions. We found that geometric normalization
with respect to the nucleus shape based on non-rigid image registration has a
significant influence on the location of the genomic regions. BACKGROUND: In mammals, one of the female X chromosomes and all imprinted genes
are expressed exclusively from a single allele in somatic cells. To evaluate
structural changes associated with allelic silencing, we have applied a recently
developed Hi-C assay that uses DNase I for chromatin fragmentation to mouse F1
hybrid systems.
RESULTS: We find radically different conformations for the two female mouse X
chromosomes. The inactive X has two superdomains of frequent intrachromosomal
contacts separated by a boundary region. Comparison with the recently reported
two-superdomain structure of the human inactive X shows that the genomic content
of the superdomains differs between species, but part of the boundary region is
conserved and located near the Dxz4/DXZ4 locus. In mouse, the boundary region
also contains a minisatellite, Ds-TR, and both Dxz4 and Ds-TR appear to be
anchored to the nucleolus. Genes that escape X inactivation do not cluster but
are located near the periphery of the 3D structure, as are regions enriched in
CTCF or RNA polymerase. Fewer short-range intrachromosomal contacts are detected
for the inactive alleles of genes subject to X inactivation compared with the
active alleles and with genes that escape X inactivation. This pattern is also
evident for imprinted genes, in which more chromatin contacts are detected for
the expressed allele.
CONCLUSIONS: By applying a novel Hi-C method to map allelic chromatin contacts,
we discover a specific bipartite organization of the mouse inactive X chromosome
that probably plays an important role in maintece of gene silencing. The long non-coding RNA Xist directs a remarkable instance of developmentally
regulated, epigenetic change known as X Chromosome Inactivation (XCI). By
spreading in cis across the X chromosome from which it is expressed, Xist RNA
facilitates the creation of a heritably silent, heterochromatic nuclear
territory that displays a three-dimensional structure distinct from that of the
active X chromosome. How Xist RNA attaches to and propagates across a chromosome
and its influence over the three-dimensional (3D) structure of the inactive X
are aspects of XCI that have remained largely unclear. Here, we discuss studies
that have made significant contributions towards answering these open questions. The mammalian inactive X chromosome (Xi) condenses into a bipartite structure
with two superdomains of frequent long-range contacts, separated by a hinge
region. Using Hi-C in edited mouse cells with allelic deletions or inversions
within the hinge, here we show that the conserved Dxz4 locus is necessary to
maintain this bipartite structure. Dxz4 orientation controls the distribution of
contacts on the Xi, as shown by a massive reversal in long-range contacts after
Dxz4 inversion. Despite an increase in CTCF binding and chromatin accessibility
on the Xi in Dxz4-edited cells, only minor changes in TAD structure and gene
expression were detected, in accordance with multiple epigenetic mechanisms
ensuring X silencing. We propose that Dxz4 represents a structural platform for
frequent long-range contacts with multiple loci in a direction dictated by the
orientation of its bank of CTCF motifs, which may work as a ratchet to form the
distinctive bipartite structure of the condensed Xi. |
Why mothers with a CYP2D6 ultrarapid metabolizer phenotype may expose their infants to risk of adverse events when taking codeine while breastfeeding? | Mothers with a CYP2D6 ultrarapid metabolizer phenotype may expose their infants to risk of adverse events when taking codeine while breastfeeding, by producing more of the active metabolite, morphine. | |
Which R package can infer protein-protein interactions via thermal proximity coaggregation (TPCA)? | Rtpca is an R package implemented methods for inferring protein-protein interactions (PPIs) based on thermal proteome profiling experiments of a single condition or in a differential setting via an approach called thermal proximity coaggregation. It offers user-friendly tools to explore datasets for their PPI predictive performance and easily integrates with available R packages. | SUMMARY: Rtpca is an R package implementing methods for inferring
protein-protein interactions (PPIs) based on thermal proteome profiling
experiments of a single condition or in a differential setting via an approach
called thermal proximity coaggregation. It offers user-friendly tools to explore
datasets for their PPI predictive performance and easily integrates with
available R packages.
AVAILABILITY AND IMPLEMENTATION: Rtpca is available from Bioconductor
(https://bioconductor.org/packages/Rtpca).
SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics
online. |
What is the mechanisms of action of Evinacumab? | Evinacumab is angiopoietin-like protein 3 (ANGPTL3) monoclonal antibody that was shown to substantially reduce low-density lipoprotein cholesterol concentration. | Angiopoietin-like 3 protein (ANGPTL3) is an inhibitor of both lipoprotein lipase
and endothelial lipase in humans. Population studies indicate a relationship
between loss of function mutations in ANGPTL3 and favorable reductions in
triglycerides and non- high-density lipoprotein cholesterol. In addition, loss
of function mutations is associated with a reduced risk of coronary artery
disease. Whereas ANGPTL3's role in human lipid metabolism has yet to be fully
clarified, it is unlikely that ANGPTL3 impacts cholesterol uptake via the
low-density lipoprotein-receptor, unlike the proprotein convertase
subtilisin/kexin9 inhibitors. In contrast to other forms of lipid-lowering
therapy, ANGPTL3 inhibition may improve insulin sensitivity. The promise of this
new therapy, particularly its independence from the low-density
lipoprotein-receptor, has prompted the creation of a monoclonal antibody
inhibitor; evinacumab. Evinacumab has shown favorable lipid-lowering action in
both human and mouse models. Efficacy trials are currently ongoing and will be
completed in the near future. In addition, ANGPTL3 inhibition via an antisense
oligonucleotide was performed in healthy human subjects, which resulted in a
dose-dependent reduction in circulating ANGPTL3 levels and an antiatherogenic
lipid profile. When tested in mouse models, administration of the antisense
oligonucleotide caused a reduction in progression of atherosclerosis. Further
investigation is required to evaluate the efficacy, safety and net benefit of
clinical ANGPTL3 inhibition before it can be accepted into clinical practice. INTRODUCTION: Cardiovascular (CV) diseases are the leading cause of death and
disability in the developed countries. Lipid-lowering therapy is a cornerstone
of the CV risk modification strategy. The first line treatment for
hyperlipidemia is statins, which decrease low-density lipoprotein cholesterol
(LDL-C) by 30-50% and proportionally reduce the CV events. However, they are not
always enough to achieve LDL-C goals in many patients, and some patients are
statin intolerant. For this reason, new powerful injectable lipid-lowering drugs
have been developed.
AREAS COVERED: The aim of this narrative review was to summarize the more recent
clinical data on safety and tolerability of injectable lipid-lowering drugs.
After an attentive literature search, the authors resumed here information on
proprotein convertase subtilisin/kexin 9 inhibitors (evolocumab and alirocumab),
small interfering RNA molecule inclisiran, antisense oligonucleotides
(mipomersen, volanesorsen, ISIS 681257), and drugs targeting angiopoietin-like
protein 3 (evinacumab, IONIS-ANGPTL3Rx).
EXPERT OPINION: Injectable lipid-lowering therapy for patients at high risk for
CV disease complications or with severe inherited hypercholesterolemias can be
an important element of the available therapeutic armamentarium. Clinical data
prove the favorable risk-benefit profile of evolocumab, alirocumab, and
inclisiran. Mipomersen, volanesorsen, ISIS 681257, evinacumab, and
IONIS-ANGPTL3Rx safety is currently less extensively studied, especially in
patients with comorbidities and polypharmacotherapy. BACKGROUND: Hypertriglyceridemia is associated with increased cardiovascular
risk and may be caused by impaired lipoprotein clearance. Angiopoietin-like
protein 3 (ANGPTL3) inhibits lipoprotein lipase activity, increasing
triglycerides and other lipids. Evinacumab, an ANGPTL3 inhibitor, reduced
triglycerides in healthy human volunteers and in homozygous familial
hypercholesterolemic individuals. Results from 2 Phase 1 studies in
hypertriglyceridemic subjects are reported here.
METHODS: Subjects with triglycerides >150 but ≤450 mg/dL and low-density
lipoprotein cholesterol ≥100 mg/dL (n=83 for single ascending dose study [SAD];
n=56 for multiple ascending dose study [MAD]) were randomized 3:1 to
evinacumab:placebo. SAD subjects received evinacumab subcutaneously at
75/150/250 mg, or intravenously at 5/10/20 mg/kg, monitored up to day 126. MAD
subjects received evinacumab subcutaneously at 150/300/450 mg once weekly,
300/450 mg every 2 weeks, or intravenously at 20 mg/kg once every 4 weeks up to
day 56 with 6 months of follow-up. The primary outcomes were incidence and
severity of treatment-emergent adverse events. Efficacy analyses included
changes in triglycerides and other lipids over time.
RESULTS: In the SAD, 32 (51.6%) versus 9 (42.9%) subjects on evinacumab versus
placebo reported treatment-emergent adverse events. In the MAD, 21 (67.7%)
versus 9 (75.0%) subjects on subcutaneously evinacumab versus placebo and 6
(85.7%) versus 1 (50.0%) on intravenously evinacumab versus placebo reported
treatment-emergent adverse events. No serious treatment-emergent adverse events
or events leading to death or treatment discontinuation were reported.
Elevations in alanine aminotransferase (7 [11.3%] SAD), aspartate
aminotransferase (4 [6.5%] SAD), and creatinine phosphokinase (2 [3.2%) SAD, 1
[14.3%] MAD) were observed with evinacumab (none in the placebo groups), which
were single elevations and were not dose-related. Dose-dependent reductions in
triglycerides were observed in both studies, with maximum reduction of 76.9% at
day 3 with 10 mg/kg intravenously (P<0.0001) in the SAD and of 83.1% at day 2
with 20 mg/kg intravenously once every 4 weeks (P=0.0003) in the MAD.
Significant reductions in other lipids were observed with most evinacumab doses
versus placebo.
CONCLUSION: Evinacumab was well-tolerated in 2 Phase 1 studies. Lipid changes in
hypertriglyceridemic subjects were similar to those observed with ANGPTL3
loss-of-function mutations. Because the latter is associated with reduced
cardiovascular risk, ANGPTL3 inhibition may improve clinical outcomes.
CLINICAL TRIAL REGISTRATION: https://www.clinicaltrials.gov. Unique identifiers:
NCT01749878 and NCT02107872. PURPOSE OF REVIEW: Homozygous familial hypercholesterolemia (HoFH) is a rare
disorder associated with early atherosclerotic disease due to impairment of the
LDL receptor (LDLR) pathway. Because of their molecular defect, current
treatment options have limited success in bringing HoFH patient to LDL-C target
and morbidity and mortality remain high. We review current and upcoming
therapies directed at HoFH, including gene therapy.
RECENT FINDINGS: Recent real-world studies have confirmed the strength in
lomitapide as a treatment adjunct to statins and other lipid-lowering therapies
in HoFH patients. The approval of proprotein convertase subtilisin/kexin type 9
(PCSK9) inhibitor monoclonal antibodies has also been a welcome addition to the
treatment armamentarium offering an additional average reduction in LDL-C levels
of 24% when added to background lipid-lowering therapies in this population.
Although achieving adequate LDL-C levels in this population is difficult, there
are several therapies on the horizon that may help more patients reach goal.
Evinacumab, a monoclonal antibody against ANGPTL3, has been shown to
substantially reduce LDL-C of an average of 49%, independently of residual LDLR
activity. RNA interference targeting PCSK9 and ANGPTL3 shows promise in clinical
trials. Adeno-associated virus-mediated gene transfer and gene editing
techniques are in early clinical and preclinical development.
SUMMARY: LDL-C lowering in HoFH patients remains very challenging. However,
novel treatment options are emerging. Upcoming therapies directed at PCSK9 and
ANPTL3 may offer additional LDL-C reduction, to help patients achieve adequate
LDL-C levels. Gene therapy and gene editing techniques, if proven effective, may
offer a unique opportunity to treat patients with a one-time treatment. INTRODUCTION: The prevalence of hypertriglyceridemia (HTG) is increasing.
Elevated triglyceride (TG) levels are associated with an increased
cardiovascular disease (CVD) risk. Moreover, severe HTG results in an elevated
risk of pancreatitis, especially in severe HTG with an up to 350-fold increased
risk. Both problems emphasize the clinical need for effective TG lowering.
AREAS COVERED: The purpose of this review is to discuss the currently available
therapies and to elaborate the most promising novel therapeutics for TG
lowering.
EXPERT OPINION: Conventional lipid lowering strategies do not efficiently lower
plasma TG levels, leaving a residual CVD and pancreatitis risk. Both
apolipoprotein C-III (apoC-III) and angiopoietin-like 3 (ANGPTL3) are important
regulators in TG-rich lipoprotein (TRL) metabolism. Several novel agents
targeting these linchpins have ended phase II/III trials. Volanesorsen targeting
apoC-III has shown reductions in plasma TG levels up to 90%. Multiple ANGPLT3
inhibitors (evinacumab, IONIS-ANGPTL3-LRx, ARO-ANG3) effectuate TG reductions up
to 70% with concomitant potent reduction in all other apoB containing
lipoprotein fractions. We expect these therapeutics to become players in the
treatment for (especially) severe HTG in the near future. PURPOSE OF REVIEW: Homozygous familial hypercholesterolemia (HoFH) is an orphan
disease caused by biallelic mutations at the LDL receptor (LDLR) gene, with a
prevalence estimated at 1 : 250 000 to 1 : 630 000. HoFH is characterized by
extremely elevated plasma levels of LDL-C greater than 10 mmol/l (>387 mg/dl),
tendinous and cutaneous xanthomas in youth and premature atherosclerotic
cardiovascular disease (ASCVD). The expected prevalence varies from country to
country depending on the presence of founder effects, genetic probability and
life expectancy. Untreated, HoFH is a fatal condition before age 30. Plasma
levels of LDL-C are the major cause of mortality and the therapeutic target.
Statin therapy led to a remarkable improvement in survival but is of limited use
in loss-of-function LDLR gene variants or 'null' mutations. Inhibitors of PCSK9
are a useful adjunct in patients with LDLR mutations with residual activity.
Extracorporeal LDL filtration has improved survival since its introduction three
decades ago.
RECENT FINDINGS: Novel therapies, not dependent on a functioning LDLR include
lomitapide and mipomersen, which decrease hepatic apolipoprotein B secretion,
and evinacumab, directed at the angiopoietin like-3 protein (ANGPLT-3).
SUMMARY: Over the past 3-4 decades, the survival of patients with HoFH has
increased markedly. New therapeutic options offer new hope. BACKGROUND: Homozygous familial hypercholesterolemia is characterized by
premature cardiovascular disease caused by markedly elevated levels of
low-density lipoprotein (LDL) cholesterol. This disorder is associated with
genetic variants that result in virtually absent (null-null) or impaired
(non-null) LDL-receptor activity. Loss-of-function variants in the gene encoding
angiopoietin-like 3 (ANGPTL3) are associated with hypolipidemia and protection
against atherosclerotic cardiovascular disease. Evinacumab, a monoclonal
antibody against ANGPTL3, has shown potential benefit in patients with
homozygous familial hypercholesterolemia.
METHODS: In this double-blind, placebo-controlled, phase 3 trial, we randomly
assigned in a 2:1 ratio 65 patients with homozygous familial
hypercholesterolemia who were receiving stable lipid-lowering therapy to receive
an intravenous infusion of evinacumab (at a dose of 15 mg per kilogram of body
weight) every 4 weeks or placebo. The primary outcome was the percent change
from baseline in the LDL cholesterol level at week 24.
RESULTS: The mean baseline LDL cholesterol level in the two groups was 255.1 mg
per deciliter, despite the receipt of maximum doses of background lipid-lowering
therapy. At week 24, patients in the evinacumab group had a relative reduction
from baseline in the LDL cholesterol level of 47.1%, as compared with an
increase of 1.9% in the placebo group, for a between-group least-squares mean
difference of -49.0 percentage points (95% confidence interval [CI], -65.0 to
-33.1; P<0.001); the between-group least-squares mean absolute difference in the
LDL cholesterol level was -132.1 mg per deciliter (95% CI, -175.3 to -88.9;
P<0.001). The LDL cholesterol level was lower in the evinacumab group than in
the placebo group in patients with null-null variants (-43.4% vs. +16.2%) and in
those with non-null variants (-49.1% vs. -3.8%). Adverse events were similar in
the two groups.
CONCLUSIONS: In patients with homozygous familial hypercholesterolemia receiving
maximum doses of lipid-lowering therapy, the reduction from baseline in the LDL
cholesterol level in the evinacumab group, as compared with the small increase
in the placebo group, resulted in a between-group difference of 49.0 percentage
points at 24 weeks. (Funded by Regeneron Pharmaceuticals; ELIPSE HoFH
ClinicalTrials.gov number, NCT03399786.). Publisher: La quilomicronemia familiar es una condición en que una mutación
genética altera la capacidad de metabolizar los triglicéridos que viajan en las
lipoproteínas, causando elevación extrema de triglicéridos plasmáticos y
complicaciones asociadas. La complicación más frecuente es la pancreatitis, que
puede llevar a falla multiorgánica o insuficiencia pancreática. La
quilomicronemia familiar también afecta la calidad de vida, las relaciones
sociales y el desarrollo profesional. El gen más frecuentemente afectado en la
quilomicronemia familiar es el de lipoproteína lipasa-1 (LPL), enzima que
hidroliza triglicéridos circulantes para su captación tisular. Mutaciones en
genes (como APOC2, APOAV, LMF-1, GPIHBP-1) que codifican para proteínas que
regulan la maduración, transporte o polimerización de lipoproteína lipasa-1,
también pueden estar involucradas. Sin embargo, en cerca del 30% de los
pacientes no se encuentra la variante causal. La quilomicronemia familiar debe
sospecharse en casos de hipertrigliceridemia extrema, resistente al tratamiento
convencional, o que se acompaña de xantomas eruptivos, lipemia retinalis o dolor
abdominal. La disponibilidad de escalas de riesgo y pruebas genéticas deben
promover la detección oportuna. La nutrición se basa en una dieta muy baja en
grasa con adecuada suplencia de vitaminas liposolubles y ácidos grasos
esenciales, además de evitar el consumo de alcohol. Si bien el tratamiento
farmacológico incluye fibratos y ácidos grasos omega 3, el enfoque actual
privilegia agentes biotecnológicos dirigidos a los defectos moleculares propios
de la enfermedad. Ello incluye un oligonucleótido antisentido dirigido contra
apoC-III (volanesorsen), un anticuerpo monoclonal contra la proteína similar a
angiopoietina tipo 3 (evinacumab), y otros compuestos en desarrollo. BACKGROUND: Patients with refractory hypercholesterolemia, who have high
low-density lipoprotein (LDL) cholesterol levels despite treatment with
lipid-lowering therapies at maximum tolerated doses, have an increased risk of
atherosclerosis. In such patients, the efficacy and safety of subcutaneous and
intravenous evinacumab, a fully human monoclonal antibody against
angiopoietin-like 3, are not known.
METHODS: In this double-blind, placebo-controlled, phase 2 trial, we enrolled
patients with or without heterozygous familial hypercholesterolemia who had
refractory hypercholesterolemia, with a screening LDL cholesterol level of 70 mg
per deciliter or higher with atherosclerosis or of 100 mg per deciliter or
higher without atherosclerosis. Patients were randomly assigned to receive
subcutaneous or intravenous evinacumab or placebo. The primary end point was the
percent change from baseline in the LDL cholesterol level at week 16 with
evinacumab as compared with placebo.
RESULTS: In total, 272 patients were randomly assigned to the following groups:
subcutaneous evinacumab at a dose of 450 mg weekly (40 patients), 300 mg weekly
(43 patients), or 300 mg every 2 weeks (39 patients) or placebo (41 patients);
or intravenous evinacumab at a dose of 15 mg per kilogram of body weight every 4
weeks (39 patients) or 5 mg per kilogram every 4 weeks (36 patients) or placebo
(34 patients). At week 16, the differences in the least-squares mean change from
baseline in the LDL cholesterol level between the groups assigned to receive
subcutaneous evinacumab at a dose of 450 mg weekly, 300 mg weekly, and 300 mg
every 2 weeks and the placebo group were -56.0, -52.9, and -38.5 percentage
points, respectively (P<0.001 for all comparisons). The differences between the
groups assigned to receive intravenous evinacumab at a dose of 15 mg per
kilogram and 5 mg per kilogram and the placebo group were -50.5 percentage
points (P<0.001) and -24.2 percentage points, respectively. The incidence of
serious adverse events during the treatment period ranged from 3 to 16% across
trial groups.
CONCLUSIONS: In patients with refractory hypercholesterolemia, the use of
evinacumab significantly reduced the LDL cholesterol level, by more than 50% at
the maximum dose. (Funded by Regeneron Pharmaceuticals; ClinicalTrials.gov
number, NCT03175367.). Introduction: Homozygous Familial Hypercholesterolemia (HoFH) is a very severe
genetic form of hypercholesterolemia. Lacking LDL receptors in the liver,
subjects with HoFH have raised plasma levels of LDL cholesterol, and up to 100
times higher risk of premature atherosclerotic cardiovascular disease than the
general population.Areas covered: This evaluation is of a phase 3 trial of
evinacumab; Evinacumab Lipid Studies in Patients with Homozygous Familial
Hypercholesterolemia (ELIPSE HoFH). Evinacumab is a human monoclonal antibody
inhibitor of angiopoietin-like protein 3 (ANGPTL3). In ELIPSE HoFH, evinacumab
reduced LDL cholesterol by 47.1 ± 4.6%, HDL cholesterol by 30.4%, and
triglycerides by 50.4 ± 7.7%.Expert opinion: Evinacumab is not the ideal
treatment for HoFH as it does not reduce LDL cholesterol levels to treatment
targets while increasing HDL cholesterol. Although the incidence of adverse
effects with evinacumab was low in ELIPSE HoFH, further studies are necessary to
clarify its effects on liver enzymes and clinical cardiovascular outcomes.
Evinacumab is a candidate to become the standard treatment for HoFH, as it may
be better tolerated and/or more efficacious than the presently available
specific treatment (lomitapide). However, the widespread use of evinacumab to
treat high triglycerides or LDL cholesterol is unlikely due to evinacumab
decreasing HDL cholesterol. |
Do honey contain diastases/amylases? | Yes
honey contain the protein amylase. | Previous experiments to separate the amylase and "invertase" of honey by
chromatography on sephadex-gels were unsuccesful. It was shown that
honey-amylase, -like amylases form other sources -- has hydrophobic properties.
Therefore it was possible to separate amylase-activity from other activities by
means of hydrophobic affinity chromatography on phenyl-butylamine-Sepharose 4B. The major alpha-amylase in honey was characterized. The optimum pH range and
temperature were determined for the enzyme as 4.6 to 5.3 and 55 degrees C,
respectively. The enzyme was stable at pH values from 7 to 8. The half-lives of
the purified enzyme at different temperatures were determined. The activation
energy for heat inactivation of honey amylase was 114.6 kJ/mol. The enzyme
exhibited Michaelis-Menten kinetics with soluble starch and gave KM and Vmax
values of 0.72 mg/mL and 0.018 units/mL, respectively. The enzyme was inhibited
by CuCl (34.3%), MgCl2 (22.4%), and HgCl2 (13.4%), while CaCl2, MnCl2, and ZnSO4
did not have any effect. Starch had a protective effect on thermal stability of
honey amylase. Therefore, it might be critical to process or control the amylase
in honey before incorporation into starch-containing foods to aid in the
preservation of starch functionality. One step could involve heat treating honey
with other ingredients, especially those that dilute and acidify the honey
environment. A new rapid method for the determination of honey diastase activity using direct
potentiometric principles has been proposed. A platinum redox sensor has been
used to quantify the amount of free triiodide released from a starch triiodide
complex after starch hydrolysis by honey diastase. The method was tested on
honey samples with varying diastase activities. The first 5 min of data for each
sample were used for linear regression analysis in order to calculate diastase
activity. The new method was compared with classical Schade and commercial
Phadebas procedures. The results showed good correlations with both methods and
offered a simple method for unit conversion to DN units for diastase activity,
making the method suitable for routine analysis. This study aimed to screen alpha-amylase producing microorganisms from honey as
a low water activity medium, a suitable source for selecting stable and
cost-beneficial bacterial enzyme production systems. Plackett-Burman method was
used to select twelve effective factors including pH, inoculum size,
temperature, time, corn starch, KH2PO4, peptone, MgSO4, CaCl2, NaCl, glycerin,
and yeast extract concentrations on bacterial alpha-amylases production yield.
The Box-Behnken method was utilized to optimize the level of selected
significant factors. The stability of bacterial alpha-amylases was also
determined in low pH and high-temperature conditions. In addition, in silico
study was used to create the alpha-amylase structure and study the stability in
high-temperature and low water available condition. Among all isolated and
characterized microorganisms, Bacillus megaterium produced the highest amount of
alpha-amylases. The in silico data showed the enzyme 3D structure similarity to
alpha-amylase from Halothermothrix orenii and highly negative charge amino acids
on its surface caused the enzyme activity and stability in low water conditions.
Based on Box-Behnken results, the temperature 35 °C, pH 6 and starch 40 g/l were
determined as the optimum level of significant factors to achieve the highest
alpha-amylases unit (101.44 U/ml). This bacterial alpha-amylases enzyme showed
stability at pH 5 and a range of temperatures from 40 to 60 °C that indicates
this enzyme may possess the potential for using in industrial processes. |
What year was the first successful human heart transplant performed? | The first human heart transplant in 1967 was performed using a deceased donor heart, | The world's first human-to-human heart transplant was performed at Groote Schuur
Hospital on the 2nd December 1967. Between 1967 and 1973, 10 patients underwent
orthotopic heart transplantation. Four lived for more than 1 year. The longest
survivor died after 12 1/2 years, and one patient remains alive and fully
employed 11 1/2 years after transplantation. Since 1974, 44 patients have
undergone heterotopic heart transplantation, whereby the donor heart is inserted
in parallel with the recipient's own heart. Four of these patients have
undergone retransplantation for acute or chronic rejection. Survival has been
almost 60% for 1 year, falling to 21% by 5 years. The major complications of
heart transplantation have been early acute and late chronic rejection;
immunosuppression has been complicated by a high incidence of infection,
particularly during the first year, and by a 10% incidence of the development of
maligt tumors. A portable hypothermic perfusion system has been developed to
store and transport donor hearts for periods of up to 24 hours. For centuries, the medical community has known that human cadavers and human
organs are invaluable to medical science for transplantations. On December 3,
1967, in Cape Town, South Africa, Dr. Christian Barnard revolutionized organ
transplantation with the first successful human heart transplant. With it came
the dawning of a new era--life for one preserved through the death of another.
But with this advancement came great demands. Patients waited, hoping sometimes
in vain, that a kidney transplant would end their long hours of suffering on
dialysis, or that a heart transplant would prolong their otherwise shortened
existence. Expectations grew, but the supply of organs did not. It became clear
that the full and continued benefit of such medical advances depended on an
increased supply of human organs for transplantation. This article focuses on
the issues surrounding the procurement of human organs for transplantation. A
brief history of transplantation and the benefits of transplantation are
presented, as well as the present means of supplying organs for transplantation
and how this supply could be enhanced by alternate procurement methods. The field of heart transplantation was built upon the discoveries of immunity
and tolerance by Landsteiner, Medawar, Burnet, and others, as well as technical
advancements in surgical technique by Carrel. Since the first successful human
heart transplant performed by Christiaan Barnard in 1967, there has been
substantial progress in the field of heart transplantation, especially over the
last several decades. With advances in immunosuppression and surgical
techniques, the rates of acute rejection and infection leading to graft failure
have declined. However, the detection of acute and chronic allograft rejection
remains one of the most important yet unsettled matters. As such, many new
horizons exist for further advancement of the field of heart transplantation and
for improving the outcomes of the patients we serve. It has been 40 years since the first human-to-human heart transplant performed
in South Africa by Christiaan Barnard in December 1967. This achievement did not
come as a surprise to the medical community but was the result of many years of
early pioneering experimental work by Alexis Carrel, Frank Mann, Norman Shumway,
and Richard Lower. Since then, refinement of donor and recipient selection
methods, better donor heart management, and advances in immunosuppression have
significantly improved survival. In this article, we hope to give a perspective
on the changing face of heart transplantation. Topics that will be covered in
this review include the changing patient population as well as recent advances
in transplantation immunology, organ preservation, allograft vasculopathy, and
immune tolerance. Christiaan (Chris) Barnard was born in 1922 and qualified in medicine at the
University of Cape Town in 1946. Following surgical training in South Africa and
the USA, Barnard established a successful open-heart surgery programme at Groote
Schuur Hospital and the University of Cape Town in 1958. In 1967, he led the
team that performed the world's first human-to-human heart transplant. The
article describing this remarkable achievement was published in the South
African Medical Journal just three weeks after the event and is one of the most
cited articles in the cardiovascular field. In the lay media as well, this first
transplant remains the most publicised event in world medical history. Although
the first heart transplant patient survived only 18 days, four of Groote Schuur
Hospital's first 10 patients survived for more than one year, two living for 13
and 23 years, respectively. This relative success amid many failures worldwide
did much to generate guarded optimism that heart transplantation would
eventually become a viable therapeutic option. This first heart transplant and
subsequent ongoing research in cardiac transplantation at the University of Cape
Town and in a few other dedicated centres over the subsequent 15 years laid the
foundation for heart transplantation to become a well-established form of
therapy for end-stage cardiac disease. During this period from 1968 to 1983,
Chris Barnard and his team continued to make major contributions to organ
transplantation, notably the development of the heterotopic ( 'piggy-back')
heart transplants; advancing the concept of brain death, organ donation and
other related ethical issues; better preservation and protection of the donor
heart (including hypothermic perfusion storage of the heart; studies on the
haemodynamic and metabolic effects of brain death; and even early attempts at
xenotransplantation. Article on the first heart transplant, performed at Groote Schuur Hospital, Cape
Town, on 3 December 1967. Reprinted from the SAMJ of 30 December 1967 to
commemorate the 50th anniversary of the transplant. The first human-to-human heart transplant was performed 50 years ago in 1967.
Heart transplantation has now entered an era of tremendous growth and
innovation. The future of heart transplantation is bright with the advent of
newer immunosuppressive medications and strategies that may even result in
tolerance. Much of this progress in heart transplant medicine is predicated on a
better understanding of acute and chronic rejection pathways through basic
science studies. The future will also include personalized medicine where
genomics and molecular science will dictate customized treatment for optimal
outcomes. The introduction of mechanical circulatory support (MCS) devices has
changed the landscape for patients with severe heart failure to stabilize the
most ill patient and make them better candidates for heart transplant. As ex
vivo preservation takes hold, we may witness an expansion of the donor pool
through the use of donation after cardiac death (DCD) donors. In addition,
further geographical donor heart sharing through ex vivo preservation may
further decrease waitlist mortality by enabling longer distance donor hearts to
be allocated for the sickest waitlist patient. It is no doubt an exciting time
to be involved in the field of heart transplantation. In this perspective, we
will summarize the present state of heart transplantation and discuss various
innovations that are being pursued. 50 years have passed since the first human to human heart transplantation,
performed by Christiaan Barnard in Cape Town December 3rd 1967. Over the years
there has been a dramatic improvement in postoperative survival, mainly due to
numerous diagnostic and therapeutic advances. Today, heart transplantation
constitutes the treatment of choice among suitable patients with severe heart
failure who worsen despite medical and surgical optimization. The world average
10-year survival has now reached more than 58 %. This text summarizes the past,
present and future. In 2017, we celebrated the 50th anniversary of the first human heart transplant
that had been carried out by the South African surgeon, Christiaan ('Chris')
Barnard at Groote Schuur Hospital in Cape Town on December 3rd, 1967. The daring
operation and the charismatic surgeon received immense public attention around
the world. The patient's progress was covered by the world's media on an almost
hourly basis. Although the patient, Mr. Louis Washansky, died after only 18
days, Barnard soon carried out a second transplant, and this patient led an
active life for almost 19 months. Remarkably, Barnard's fifth and sixth patients
lived for almost 13 and 24 years, respectively. Barnard subsequently introduced
the operation of heterotopic heart transplantation in which the donor heart
acted as an auxiliary pump, with some advantages in that early era. It took
great courage to carry out the first heart transplant, and this is why Barnard
is remembered as a pioneer in cardiac surgery. The gold standard and sole curative therapy for advanced stage heart failure is
cardiac transplantation. As the population ages, the number of patients
diagnosed with advanced heart failure and listed for transplant steadily
increases annually. However, there remains a paucity of eligible donation after
brain death (DBD) donor hearts which severely limits access to cardiac
transplantation and leads to increasing wait-list times and avoidable patient
mortalities. Though the first human heart transplant in 1967 was performed using
a deceased donor heart, the advent of brain death criteria and the ability to
avoid long warm ischemic times led donation after cardiac death (DCD)
transplantation to fall out of favor. Due the current state of cardiac
transplantation, there has been a resurgence in interest in DCD heart
transplantation leading to the development of DCD heart transplantation programs
in the UK and Australia after positive reports of successful DCD cardiac
transplantation in the pediatric literature. These programs have demonstrated
favorable post-transplantation outcomes equivalent to matched traditional DBD
transplants with current techniques and strict donor criteria. This technique
has been proven safe with favorable outcomes and has been demonstrated to
significantly increase transplant volumes and decrease patient mortality. Given
these outcomes and the high patient benefit to risk ratio, DCD donor heart
transplantation is necessary to expand the donor pool and decrease patient
mortality and should be developed in high volume experienced cardiac transplant
centers. The first successful human heart transplantation was reported on 3 December
1967, by Christiaan Barnard in South Africa. Since then this life-saving
procedure has been performed in over 120 000 patients. A limitation to the
performance of this procedure is the availability of donor hearts with as many
as 20% of patients dying before a donor's heart is available for transplant.
Today, hearts for transplantation are procured from individuals experiencing
donation after brain death (DBD). Interestingly, this, however, was not always
the case as the first heart transplants occurred after circulatory death.
Revisiting the availability of hearts for transplant from those
experiencing donation after circulatory death (DCD) could further expand the
number of hearts suitable for transplantation. There are several considerations
pertinent to transplanting hearts from those undergoing circulatory death. In
this review, we summarize the main distinctions between DBD and DCD heart
donation and discuss the research relevant to increasing the number of hearts
available for transplantation by including individual's hearts that experience
circulatory death. |
Which disease is monitored in the BIOCURA cohort? | Rheumatoid Arthritis (RA) is one of the diseases that is monitored in the BIOCURA cohort. There are other diseases that are monitored as well, such as breast cancer, ovarian cancer, and thyroid cancer. | BACKGROUND: In rheumatoid arthritis, prediction of response to TNF-alpha
inhibitor (TNFi) treatment would be of clinical value. This study aims to
discover miRNAs that predict response and aims to replicate results of two
previous studies addressing this topic.
METHODS: From the observational BiOCURA cohort, 40 adalimumab- (ADA) and 40
etanercept- (ETN) treated patients were selected to enter the discovery cohort
and baseline serum profiling on 758 miRNAs was performed. The added value of
univariately selected miRNAs (p < 0.05) over clinical parameters in prediction
of response was determined by means of the area under the receiver operating
characteristic curve (AUC-ROC). Validation was performed by TaqMan single qPCR
assays in 40 new patients.
RESULTS: Expression of miR-99a and miR-143 predicted response to ADA, and
miR-23a and miR-197 predicted response to ETN. The addition of miRNAs increased
the AUC-ROC of a model containing only clinical parameters for ADA (0.75 to
0.97) and ETN (0.68 to 0.78). In validation, none of the selected miRNAs
significantly predicted response. miR-23a was the only overlapping miRNA
compared to the two previous studies, however inversely related with response in
one of these studies. The reasons for the inability to replicate previously
proposed miRNAs predicting response to TNFi and replicate those from the
discovery cohort were investigated and discussed.
CONCLUSIONS: To date, no miRNA consistently predicting response to TNFi therapy
in RA has been identified. Future studies on this topic should meet a minimum of
standards in design that are addressed in this study, in order to increase the
reproducibility. OBJECTIVE: In rheumatoid arthritis (RA), it is of major importance to identify
non-responders to tumour necrosis factor-α inhibitors (TNFi) before starting
treatment, to prevent a delay in effective treatment. We developed a protein
score for the response to TNFi treatment in RA and investigated its predictive
value.
METHOD: In RA patients eligible for biological treatment included in the BiOCURA
registry, 53 inflammatory proteins were measured using xMAP® technology. A
supervised cluster analysis method, partial least squares (PLS), was used to
select the best combination of proteins. Using logistic regression, a predictive
model containing readily available clinical parameters was developed and the
potential of this model with and without the protein score to predict European
League Against Rheumatism (EULAR) response was assessed using the area under the
receiving operating characteristics curve (AUC-ROC) and the net reclassification
index (NRI).
RESULTS: For the development step (n = 65 patient), PLS revealed 12 important
proteins: CCL3 (macrophage inflammatory protein, MIP1a), CCL17 (thymus and
activation-regulated chemokine), CCL19 (MIP3b), CCL22 (macrophage-derived
chemokine), interleukin-4 (IL-4), IL-6, IL-7, IL-15, soluble cluster of
differentiation 14 (sCD14), sCD74 (macrophage migration inhibitory factor),
soluble IL-1 receptor I, and soluble tumour necrosis factor receptor II. The
protein score scarcely improved the AUC-ROC (0.72 to 0.77) and the ability to
improve classification and reclassification (NRI = 0.05). In validation
(n = 185), the model including protein score did not improve the AUC-ROC (0.71
to 0.67) or the reclassification (NRI = -0.11).
CONCLUSION: No proteomic predictors were identified that were more suitable than
clinical parameters in distinguishing TNFi non-responders from responders before
the start of treatment. As the results of previous studies and this study are
disparate, we currently have no proteomic predictors for the response to TNFi. BACKGROUND: Several studies have employed microarray-based profiling to predict
response to tumor necrosis factor-alpha inhibitors (TNFi) in rheumatoid
arthritis (RA); yet efforts to validate these targets have failed to show
predictive abilities acceptable for clinical practice.
METHODS: The eighty most extreme responders and nonresponders to TNFi therapy
were selected from the observational BiOCURA cohort. RNA sequencing was
performed on mRNA from peripheral blood mononuclear cells (PBMCs) collected
before initiation of treatment. The expression of pathways as well as individual
gene transcripts between responders and nonresponders was investigated.
Promising targets were technically replicated and validated in n = 40 new
patients using qPCR assays.
RESULTS: Before therapy initiation, nonresponders had lower expression of
pathways related to interferon and cytokine signaling, while also showing higher
levels of two genes, GPR15 and SEMA6B (p = 0.02). The two targets could be
validated, however, additional analyses revealed that GPR15 and SEMA6B did not
independently predict response, but were rather dose-dependent markers of
smoking (p < 0.0001).
CONCLUSIONS: The study did not identify new transcripts ready to use in clinical
practice, yet GPR15 and SEMA6B were recognized as candidate explanatory markers
for the reduced treatment success in RA smokers. |
Which gene is associated with response to abacavir? | Large studies established the effectiveness of prospective HLA-B*57:01 screening to prevent HSRs to abacavir. | Many pharmacogenomic biomarkers (PGBM) were identified and translated into
clinical practice, affecting the usage of drugs via label updates. In this
context, abacavir is one of the most brilliant examples of pharmacogenetic
studies translated into clinical practice. Pharmacogenetic studies have revealed
that abacavir HSRs are highly associated with the major histocompatibility
complex class I. Large studies established the effectiveness of prospective
HLA-B*57:01 screening to prevent HSRs to abacavir. Accordingly to these results
the abacavir label has been modified: the European Medicines Agency (EMA) and
the FDA recommend/suggested that the administration of abacavir must be preceded
by a specific genotyping test. The HLA locus is extremely polymorphic,
exhibiting many closely related alleles, making it difficult to discriminate
HLA-B*57:01 from other related alleles, and a number of different molecular
techniques have been developed recently to detect the presence of HLA-B*57:01.
In this review, we provide a summary of the available techniques used by
laboratories to genotype HLA-B*57:01, outlining the scientific and
pharmacoeconomics pros and cons. |
Is adenosine signaling prognostic for cancer outcome? | Yes, adenosine signaling has been shown to be prognostic for cancer outcome. | |
Which drugs are included in the CNIC polypill? | CNIC polypill includes atorvastatin 40mg, ramipril 10mg and aspirin 100mg. | Aim: To determine the effectiveness of Centro Nacional de Investigaciones
Cardiovasculares (CNIC)-polypill (acetylsalicylic acid 100 mg, ramipril 5/10 mg,
simvastatin 40 mg) in achieving blood pressure (BP) goals. Patients & methods: A
multicenter, observational, one cohort, prospective study. BP targets were
analyzed in patients with cardiovascular disease after 12-months treatment with
the CNIC polypill. Results: A total of 572 patients (59.4 ± 13.9 years, 57.3%
men) were analyzed. At baseline, BP was 147.1 ± 18.1/88.3 ± 10.6 mmHg, 97.1% of
patients were taken renin-angiotensin system inhibitors, 5.4% calcium
antagonists, 1.9% diuretics and 13.1% β-blockers. The proportion of patients who
achieved BP targets increased from 20.1 to 55.4% (p < 0.001). Conclusion: In
routine practice, switching from usual care to the CNIC-polypill in patients
with cardiovascular disease could facilitate achieving BP goals. INTRODUCTION AND OBJECTIVES: To compare the pharmacodynamics of the CNIC
polypill (atorvastatin 40mg/ramipril 10mg/aspirin 100mg) in terms of low-density
lipoprotein cholesterol (LDL-C) and systolic blood pressure (SBP), with the
corresponding reference products (atorvastatin and ramipril).
METHODS: This was a multicenter, randomized, open-label, and parallel 3-arm
study comparing the effect of the CNIC polypill vs ramipril 10mg and
atorvastatin 40mg on SBP and LDL-C. The coprimary endpoints were differences in
the adjusted mean 24-hour SBP (using ambulatory BP measurement) and LDL-C during
the study period estimated using an ANCOVA model.
RESULTS: Of the 241 patients included in the per protocol population, 84
received the CNIC polypill (group A), 84 atorvastatin (group B), and 73 ramipril
(group C). SBP decreased from 139.3±12.5 to 133.2±12.9mmHg in group A and from
138.1±11.9 to 134.0±12.8mmHg in group C (baseline adjusted mean difference for
the decrease in SBP was 1.77mmHg (90%CI, -0.5 to 4.0) in favor of group A,
without reaching statistical significance. LDL-C was reduced by 33.9±21.6 and
29.2±25.8mg/dL in groups A and B, respectively (baseline adjusted mean
difference for the decrease in LDL-C was 7.0% (90%CI, 1.5-12.4), a significantly
greater decrease with the polypill). The 3 treatments were well tolerated.
CONCLUSIONS: The results of this study rule out a negative effect on blood
pressure of the interaction between the components of the CNIC polypill. The
reduction in LDL-C was greater in the CNIC polypill group, suggesting a
synergistic effect of the components. |
Which human tissue synthesize CRP? | CRP is predominantly produced in the liver in a native pentameric form (nCRP). | In conditions of acute and chronic inflammation hepatic detoxification capacity
is severely impaired due to coordinated downregulation of drug metabolizing
enzymes and transporters. Using global transcriptome analysis of liver tissue
from donors with pathologically elevated C-reactive protein (CRP), we observed
comparable extent of positive and negative acute phase response, where the top
upregulated gene sets included immune response and defense pathways while
downregulation occurred mostly in metabolic and catabolic pathways including
many important drug metabolizing enzymes and transporters. We hypothesized that
microRNAs (miRNA), which usually act as negative regulators of gene expression,
contribute to this process. Microarray and quantitative real-time PCR analyses
identified differentially expressed miRNAs in liver tissues from donors with
elevated CRP, cholestasis, steatosis, or non-alcoholic steatohepatitis. Using
luciferase reporter constructs harboring native and mutated 3'-untranslated gene
regions, several predicted miRNA binding sites on RXRα (miR-130b-3p), CYP2C8
(miR-452-5p), CYP2C9 (miR-155-5p), CYP2C19 (miR-155-5p, miR-6807-5p), and CYP3A4
(miR-224-5p) were validated. HepaRG cells transfected with miRNA mimics showed
coordinate reductions in mRNA levels and several cytochrome P450 enzyme
activities particularly for miR-155-5p, miR-452-5p, and miR-6807-5p, the only
miRNA that was deregulated in all four pathological conditions. Furthermore we
observed strong negative correlations between liver tissue miRNA levels and
hepatic CYP phenotypes. Since miR-155 is well known for its multifunctional
roles in immunity, inflammation, and cancer, our data suggest that this and
other miRNAs contribute to coordinated downregulation of drug metabolizing
enzymes and transporters in inflammatory conditions. |
What is the relationship between the X chromosome and a neutrophil drumstick? | In particular, up to 17% of neutrophil nuclei of healthy women exhibit a drumstick-shaped appendage that contains the inactive X chromosome. | The sex chromosomal constitution has been determined in various types of human
leukocytes at interphase by use of fluorescence in situ hybridization with X-
and/or Y-specific DNA probes. It is found that during aging and differentiation
of myelocytes into polymorphs there is no significant change in the relative
frequency of various types of male and female cells with a specific type of sex
chromosomal constitution. Non-random variability of the relative proximity
between the X chromosomes within the nuclei is also observed in female cells.
Moreover, we are the first to determine that sex-specific "drumsticks" and
"sessile nodules" in female polymorphs originate from the X chromosomes and that
non-sex-specific "drumstick-like" bodies in male polymorphs are of Y chromosomal
origin. An X chromosome specific nucleic acid probe was used to study the positions of
the X chromosomes in leukocyte nuclei by in situ hybridization to smears of
peripheral blood. This autoradiographic approach allowed the first direct
demonstration of the presence of X chromosomal material in the drumstick-like
structures of female polymorphonuclear leukocytes. An individual with normal male habitus, body proportions, and secondary sexual
characteristics was admitted to the hospital with head trauma. A routine blood
smear demonstrated that 36% of the granulocytes had "drumsticks". Chromosomal
analysis revealed a 46,XYqh+ karyotype. the extremely large Y chromosome was
located by quinacrine fluorescence in the "drumstick" of the polymorphonuclear
granulocytes. The presence of a large Y chromosome may thus produce
pseudo-drumsticks. Fluorescent staining can distinguish between true drumsticks
bearing the inactive X of normal females and the pseudo-drumsticks in a normal
male produced by a large Y chromosome. The nuclei of human neutrophils typically consist of a linear array of three or
four lobes joined by DNA-containing filaments. Terminal lobes are connected to
internal lobes via a single filament, while internal lobes have two filaments,
each to an adjacent lobe. Some lobes also have appendages of various shapes and
sizes. In particular, up to 17% of neutrophil nuclei of healthy women exhibit a
drumstick-shaped appendage that contains the inactive X chromosome. This report
provides a detailed analysis of the relationship between nuclear morphology and
the location of the X and Y chromosomes in human neutrophils. Fluorescent in
situ hybridization analysis revealed that the X and the Y chromosomes of male
neutrophil nuclei are randomly distributed among nuclear lobes. Similarly, in
female neutrophil nuclei with a drumstick appendage, the active X chromosome is
also randomly distributed among lobes. In contrast, the inactive X chromosome is
preferentially located in a terminal lobe in over 90% nuclei with drumsticks.
Within the terminal lobe of nuclei with drumsticks, the inactive X chromosome
lies distal to the point of filament attachment in 80% of the nuclei. The
inactive X chromosome also exhibits a specific orientation within the drumstick
appendage, with over 95% of nuclei having the X centromere located toward the
tip of the appendage. Female nuclei without a drumstick appendage also have one
of the X chromosomes (presumably the inactive chromosome) preferentially
situated in a terminal lobe. Nonrandom distribution of the inactive X chromosome
is discussed in the context of a model that considers chromosomes as
determits of neutrophil nuclear morphology. Granulocytic early progenitors and terminally differentiated - mature
granulocytes with segmented nuclei were studied using computer-assisted diameter
and heterochromatin optical image densitometry to provide more information on
the nuclear size and heterochromatin condensation state. Bone marrow smears of
patients suffering from chronic myeloid leukaemia untreated as well as treated
with "specific" anti-leukaemic therapy with imatinib mesylate are a convenient
model for such study because they possess a satisfactory number of cells for
diameter and optical density measurements. In addition, the identification of
developmental stages of granulocytes is very easy and the morphology is not
different from that in not-leukaemic persons. As it was expected, the mean
diameter of nuclear segments in fully differentiated and mature granulocytes was
much smaller than that in non-segmented nuclei of early granulocytic precursors.
Therefore, no wonder that the heterochromatin condensation state in nuclear
segments of mature granulocytes was much larger than in non-segmented nuclei of
granulocytic progenitors. On the other hand, the sum of mean diameters of all
nuclear segments per cell was close to the mean nuclear diameter of early
granulocytic progenitors. The heterochromatin condensation state in granulocytic
progenitors or fully differentiated mature granulocytes exhibited marked
stability and did not change after the anti-leukaemic therapy. In addition, Barr
bodies of characteristic drumstick appearance bearing inactive X chromosome in
interphase nuclei of mature granulocytes in fertile female patients exhibited a
heterochromatin condensation state similar to nuclear segments. This
heterochromatin condensation state was also stable and constant, and was not
apparently influenced by the anti-leukaemic therapy. |
The formation of which inflammatory molecule is regulated by MAP3K8 (TPL2)? | MAP3K8 (Tpl2) regulates the formation of inflammatory molecule IL-1β | OBJECTIVE: Activation of extracellular signal-regulated kinase-(ERK)-1/2 by
cytokines in adipocytes is involved in the alterations of adipose tissue
functions participating in insulin resistance. This study aims at identifying
proteins regulating ERK1/2 activity, specifically in response to inflammatory
cytokines, to provide new insights into mechanisms leading to abnormal adipose
tissue function.
RESEARCH DESIGN AND METHODS: Kinase activities were inhibited with
pharmacological inhibitors or siRNA. Lipolysis was monitored through glycerol
production. Gene expression in adipocytes and adipose tissue of obese mice and
subjects was measured by real-time PCR.
RESULTS: IkappaB kinase-(IKK)-beta inhibition prevented mitogen-activated
protein (MAP) kinase kinase (MEK)/ERK1/2 activation in response to interleukin
(IL)-1beta and tumor necrosis factor (TNF)-alpha but not insulin in 3T3-L1 and
human adipocytes, suggesting that IKKbeta regulated a MAP kinase kinase kinase
(MAP3K) involved in ERK1/2 activation induced by inflammatory cytokines. We show
that the MAP3K8 called Tpl2 was expressed in adipocytes and that IL-1beta and
TNF-alpha activated Tpl2 and regulated its expression through an IKKbeta
pathway. Pharmacological inhibition or silencing of Tpl2 prevented MEK/ERK1/2
activation by these cytokines but not by insulin, demonstrating its involvement
in ERK1/2 activation specifically in response to inflammatory stimuli.
Importantly, Tpl2 was implicated in cytokine-induced lipolysis and in insulin
receptor substrate-1 serine phosphorylation. Tpl2 mRNA expression was
upregulated in adipose tissue of obese mice and patients and correlated with
TNF-alpha expression.
CONCLUSIONS: Tpl2 is selectively involved in inflammatory cytokine-induced
ERK1/2 activation in adipocytes and is implicated in their deleterious effects
on adipocyte functions. The deregulated expression of Tpl2 in adipose tissue
suggests that Tpl2 may be a new actor in adipose tissue dysfunction in obesity. Tumor progression locus 2 (Tpl2, also known as Map3k8 and Cot) is a
serine-threonine kinase critical in innate immunity, linking toll-like receptors
(TLRs) to TNF production through its activation of ERK. Tpl2(-/-) macrophages
have abrogated TNF production but overproduce IL-12 in response to TLR ligands.
Despite enhanced IL-12 production, Tpl2(-/-) T cells have impaired IFN-gamma
production. Therefore, the role of Tpl2 in a bona fide bacterial infection where
all of these cytokines are important in host defense is unclear. To address this
issue, we infected Tpl2(-/-) mice with the model pathogen Listeria
monocytogenes. We found that Tpl2(-/-) mice infected i.v. with L. monocytogenes
had increased pathogen burdens compared with wild-type mice and rapidly
succumbed to infection. Enhanced susceptibility correlated with impaired
signaling through TLR2 and nucleotide-binding oligomerization domain 2, two
receptors previously shown to mediate Listeria recognition. Surprisingly, TNF
production in response to infection was not significantly impaired, even though
Tpl2 has been implicated in the regulation of TNF. We found that the role of
Tpl2 has cell-type specific effects in regulating TNF and transduces signals
from some, but not all, pattern recognition receptors (PRR). In contrast to the
cell-type- and receptor-specific regulation of TNF, we found that Tpl2 is
essential for IL-1beta production from both macrophages and dendritic cells.
These studies implicate Tpl2 as an important mediator for collaboration of
pattern recognition receptors with danger-associated molecular patterns to
induce TNF and IL-1beta production and optimal host defense. Cot/tpl2 (also known as MAP3K8) has emerged as a new and potentially interesting
therapeutic anti-inflammatory target. Here, we report the first study of
Cot/tpl2 involvement in acute peripheral inflammation in vivo. Six hours after
an intraplantar injection of zymosan, Cot/tpl2(-/-) mice showed a 47% reduction
in myeloperoxidase activity, concomitant with a 46% lower neutrophil recruitment
and a 40% decreased luminol-mediated bioluminescence imaging in vivo.
Accordingly, Cot/tpl2 deficiency provoked a 25-30% reduction in luminol-mediated
bioluminescence and neutrophil recruitment together with a 65% lower macrophage
recruitment 4 h following zymosan-induced peritonitis. Significantly impaired
levels of G-CSF and GM-CSF and of other cytokines such as TNFα, IL-1β, and IL-6,
as well as some chemokines such as MCP-1, MIP-1β, and keratinocyte-derived
chemokine, were detected during the acute zymosan-induced intraplantar
inflammatory response in Cot/tpl2(-/-) mice. Moreover, Cot/tpl2 deficiency
dramatically decreased the production of the hypernociceptive ligand NGF at the
inflammatory site during the course of inflammation. Most importantly, Cot/tpl2
deficiency significantly reduced zymosan-induced inflammatory hypernociception
in mice, with a most pronounced effect of a 50% decrease compared with wild type
(WT) at 24 h following intraplantar injection of zymosan. At this time,
Cot/tpl2(-/-) mice showed significantly reduced NGF, TNFα, and prostaglandin
E(2) levels compared with WT littermates. In conclusion, our study demonstrates
an important role of Cot/tpl2 in the NGF, G-CSF, and GM-CSF production and
myeloperoxidase activity in the acute inflammatory response process and its
implication in inflammatory hypernociception. Interleukin-22 (IL-22), one of the cytokines secreted by T-helper 17 (Th17)
cells, binds to a class II cytokine receptor containing an IL-22 receptor 1
(IL-22R1) and IL-10R2 and influences a variety of immune reactions. IL-22 has
also been shown to modulate cell cycle and proliferation mediators such as
extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK),
but little is known about the underlying molecular mechanisms of IL-22 in
tumorigenesis. In this paper, we propose that IL-22 has a crucial role to play
in controlling epithelial cell proliferation and tumorigenesis in the breast.
IL-22 increased MAP3K8 phosphorylation through IL-22R1, followed by the
induction of MEK-ERK, JNK-c-Jun, and STAT3 signaling pathways. Furthermore,
IL-22-IL-22R1 signaling pathway activated activator protein-1 and HER2 promoter
activity. In addition, Pin1 was identified as a key positive regulator for the
phosphorylation-dependent MEK, c-Jun and STAT3 activity induced by IL-22.
Pin1(-/-) mouse embryonic fibroblasts (MEF) exhibited significantly a decrease
in IL-22-induced MEK1/2, c-Jun, and STAT3 phosphorylation compared with
Pin1(+/+) MEF. In addition, a knockdown of Pin1 prevented phosphorylation
induced by IL-22. The in vivo chorioallantoic membrane assay also showed that
IL-22 increased tumor formation of JB6 Cl41 cells. Moreover, the knockdown of
MAP3K8 and Pin1 attenuated tumorigenicity of MCF7 cells. Consistent with these
observations, IL-22 levels positively correlate with MAP3K8 and Pin1 expression
in human breast cancer. Overall, our findings point to a critical role for the
IL-22-induced MAP3K8 signaling pathway in promoting cancer-associated
inflammation in the tumor microenvironment. Chronic low-grade inflammation in adipose tissue often accompanies obesity,
leading to insulin resistance and increasing the risk for metabolic diseases.
MAP3K8 (TPL2/COT) is an important signal transductor and activator of
pro-inflammatory pathways that has been linked to obesity-induced adipose tissue
inflammation. We used human adipose tissue biopsies to study the relationship of
MAP3K8 expression with markers of obesity and expression of pro-inflammatory
cytokines (IL-1β, IL-6 and IL-8). Moreover, we evaluated obesity-induced adipose
tissue inflammation and insulin resistance in mice lacking MAP3K8 and WT mice on
a high-fat diet (HFD) for 16 weeks. Individuals with a BMI >30 displayed a
higher mRNA expression of MAP3K8 in adipose tissue compared to individuals with
a normal BMI. Additionally, high mRNA expression levels of IL-1β, IL-6 and IL-8,
but not TNF -α, in human adipose tissue were associated with higher expression
of MAP3K8. Moreover, high plasma SAA and CRP did not associate with increased
MAP3K8 expression in adipose tissue. Similarly, no association was found for
MAP3K8 expression with plasma insulin or glucose levels. Mice lacking MAP3K8 had
similar bodyweight gain as WT mice, yet displayed lower mRNA expression levels
of IL-1β, IL-6 and CXCL1 in adipose tissue in response to the HFD as compared to
WT animals. However, MAP3K8 deficient mice were not protected against
HFD-induced adipose tissue macrophage infiltration or the development of insulin
resistance. Together, the data in both human and mouse show that MAP3K8 is
involved in local adipose tissue inflammation, specifically for IL-1β and its
responsive cytokines IL-6 and IL-8, but does not seem to have systemic effects
on insulin resistance. OBJECTIVE: IBD is characterised by dysregulated intestinal immune homeostasis
and cytokine secretion. In the intestine, properly regulating pattern
recognition receptor (PRR)-mediated signalling and cytokines is crucial given
the ongoing host-microbial interactions. TPL2 (MAP3K8, COT) contributes to
PRR-initiated pathways, yet the mechanisms for TPL2 signalling contributions in
primary human myeloid cells are incompletely understood and its role in
intestinal myeloid cells is poorly defined. Furthermore, functional consequences
for the IBD-risk locus rs1042058 in TPL2 are unknown.
METHODS: We analysed protein, cytokine and RNA expression, and signalling in
human monocyte-derived macrophages (MDMs) through western blot, ELISA, real-time
PCR and flow cytometry.
RESULTS: PRR-induced cytokine secretion was increased in MDMs from rs1042058
TPL2 GG risk individuals. TPL2 activation by the Crohn's disease-associated PRR
nucleotide-oligomerisation domain (NOD)2 required PKC, and IKKβ, IKKα and IKKγ
signalling. TPL2, in turn, significantly enhanced NOD2-induced ERK, JNK and NFκB
signalling. We found that another major mechanism for the TPL2 contribution to
NOD2 signalling was through ERK-dependent and JNK-dependent caspase-1 and
caspase-8 activation, which in turn, led to early autocrine interleukin (IL)-1β
and IL-18 secretion and amplification of long-term cytokines. Importantly,
Salmonella typhimurium-induced cytokines from human intestinal myeloid-derived
cells required TPL2 as well as autocrine IL-1β and IL-18. Finally, rs1042058 GG
risk carrier MDMs from healthy individuals and patients with Crohn's disease had
increased TPL2 expression and NOD2-initiated TPL2 phosphorylation, ERK, JNK and
NFκB activation, and early autocrine IL-1β and IL-18 secretion.
CONCLUSIONS: Taken together, the rs1042058 GG IBD-risk polymorphism in TPL2
results in a gain-of-function by increasing TPL2 expression and signalling,
thereby amplifying PRR-initiated outcomes. Dendritic cells (DCs) constantly sample peripheral tissues for antigens, which
are subsequently ingested to derive peptides for presentation to T cells in
lymph nodes. To do so, DCs have to traverse many different tissues with varying
oxygen tensions. Additionally, DCs are often exposed to low oxygen tensions in
tumors, where vascularization is lacking, as well as in inflammatory foci, where
oxygen is rapidly consumed by inflammatory cells during the respiratory burst.
DCs respond to oxygen levels to tailor immune responses to such low-oxygen
environments. In the present study, we identified a mechanism of
hypoxia-mediated potentiation of release of tumor necrosis factor α (TNF-α), a
pro-inflammatory cytokine with important roles in both anti-cancer immunity and
autoimmune disease. We show in human monocyte-derived DCs (moDCs) that this
potentiation is controlled exclusively via the p38/mitogen-activated protein
kinase (MAPK) pathway. We identified MAPK kinase kinase 8 (MAP3K8) as a target
gene of hypoxia-induced factor (HIF), a transcription factor controlled by
oxygen tension, upstream of the p38/MAPK pathway. Hypoxia increased expression
of MAP3K8 concomitant with the potentiation of TNF-α secretion. This
potentiation was no longer observed upon siRNA silencing of MAP3K8 or with a
small molecule inhibitor of this kinase, and this also decreased p38/MAPK
phosphorylation. However, expression of DC maturation markers CD83, CD86, and
HLA-DR were not changed by hypoxia. Since DCs play an important role in
controlling T-cell activation and differentiation, our results provide novel
insight in understanding T-cell responses in inflammation, cancer, autoimmune
disease and other diseases where hypoxia is involved. Endogenous noradrenaline (NA) has multiple bioactive functions and, in the
central nervous system (CNS), has been implicated in modulating
neuroinflammation via β-adrenergic receptors (β-ARs). Microglia, resident
macrophages in the CNS, have a central role in the brain immune system and have
been reported to be activated by NA. However, intracellular signaling mechanisms
of the AR-mediated proinflammatory responses of microglia are not fully
understood. Using a rapid and stable in vitro reporter assay system to evaluate
IL-1β production in microglial BV2 cells, we found that NA and the β-AR agonist
isoproterenol upregulated the IL-1β reporter activity. This effect was
suppressed by β-AR antagonists. We further examined the involvement of EPAC
(exchange protein directly activated by cAMP) and TPL2 (tumor progression locus
2, MAP3K8) and found that inhibitors for EPAC and TPL2 reduced AR
agonist-induced IL-1β reporter activity. These inhibitors also suppressed
NA-induced endogenous Il1b mRNA expression and IL-1β protein production. Our
results suggest that EPAC and TPL2 are involved in β-AR-mediated IL-1β
production in microglial cells, and extend our understanding of its
intracellular signaling mechanism. |
Which company produces the Oncomine Dx target test? | The Oncomine Dx Target Test Panel is produced by Thermo Fisher Scientific. | Author information:
(1)HM Sanchinarro University Hospital-CIBERONC, Madrid, Spain.
(2)HM Sanchinarro University Hospital, Madrid, Spain.
(3)La Paz University Hospital, Madrid, Spain.
(4)Ramon y Cajal University Hospital, IRYCIS and CIBERESP, Madrid, Spain.
(5)Germans Trias i Pujol University Hospital, Badalona, Spain.
(6)Catalan Institute of Oncology-Germans Trias i Pujol University Hospital,
Universitat Autònoma Barcelona (UAB), Badalona-Applied Research Group of
Oncology (B-ARGO), Badalona, Spain.
(7)General University Hospital-ISABIAL, Alicante, Spain.
(8)Institute of Health Research-Jimenez Diaz Foundation-CIBERONC, Madrid, Spain.
(9)Institute of Health Research-Jimenez Diaz Foundation, Madrid, Spain.
(10)Vall d'Hebron University Hospital, Barcelona, Spain.
(11)Quironsalud Hospital, Barcelona, Spain.
(12)La Fe University Hospital, Valencia, Spain.
(13)Clinico San Carlos University Hospital, Madrid, Spain.
(14)Puerta del Mar University Hospital, Cadiz, Spain.
(15)Clinico de Santiago University Hospital, Santiago De Compostela, Spain.
(16)Insular Materno-Infantil University Hospital Complex, Las Palmas de Gran
Canaria, Spain.
(17)Clinic Hospital, Barcelona, Spain.
(18)Alvaro Cunqueiro Hospital, Vigo, Spain.
(19)University of Navarra Clinic, Pamplona, Spain.
(20)Marques de Valdecilla University Hospital, Santander, Spain.
(21)Hospital del Mar, Barcelona, Spain.
(22)Clinico University Hospital, Valencia, Spain.
(23)Cruces University Hospital, Baracaldo, Spain.
(24)Miguel Servet University Hospital, Zaragoza, Spain.
(25)University Hospital of Gran Canaria Doctor Negrin, Las Palmas de Gran
Canaria, Spain.
(26)12 de Octubre University Hospital, Madrid, Spain.
(27)Ramon y Cajal University Hospital, Madrid, Spain.
(28)12 de Octubre University Hospital-CIBERONC, Madrid, Spain.
(29)Ramon y Cajal University Hospital-CIBERONC, Madrid, Spain.
(30)HM Sanchinarro University Hospital-CIBERONC, Madrid, Spain. Electronic
address: [email protected]. |