Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(3)2020 02 05.
Article in English | MEDLINE | ID: mdl-32033454

ABSTRACT

Transforming growth factor ß (TGFß) is a pluripotent cytokine and regulates a myriad of biological processes. It has been established that TGFß potently inhibits skeletal muscle differentiation; however, the molecular mechanism is not clearly defined. Previously, we reported that inhibition of the TGFß canonical pathway by an inhibitory Smad, Smad7, does not reverse this effect on differentiation, suggesting that activation of receptor Smads (R-Smads) by TGFß is not responsible for repression of myogenesis. In addition, pharmacological blockade of Smad3 activation by TGFß did not reverse TGFß's inhibitory effect on myogenesis. In considering other pathways, we observed that TGFß potently activates MEK/ERK, and a pharmacological inhibitor of MEK reversed TGFß's inhibitory effect on myogenesis, as indicated by a myogenin promoter-reporter gene, sarcomeric myosin heavy chain accumulation, and phenotypic myotube formation. Furthermore, we found that c-Jun, a known potent repressor of myogenesis, which is coincidently also a down-stream target of MEK/ERK signaling, was phosphorylated and accumulates in the nucleus in response to TGFß activation. Taken together, these observations support a model in which TGFß activates a MEK/ERK/c-Jun pathway to repress skeletal myogenesis, maintaining the pluripotent undifferentiated state in myogenic progenitors.


Subject(s)
Cell Differentiation/physiology , MAP Kinase Signaling System/physiology , Muscle Development/physiology , Signal Transduction/physiology , Smad Proteins/metabolism , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism , Cell Nucleus/metabolism , Cytokines/metabolism , Humans , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/physiology , Myogenin/metabolism , Phosphorylation/physiology , Stem Cells/physiology , Trans-Activators/metabolism
2.
Hum Mol Genet ; 26(4): 753-767, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28040729

ABSTRACT

Repression of repetitive elements is crucial to preserve genome integrity and has been traditionally ascribed to constitutive heterochromatin pathways. FacioScapuloHumeral Muscular Dystrophy (FSHD), one of the most common myopathies, is characterized by a complex interplay of genetic and epigenetic events. The main FSHD form is linked to a reduced copy number of the D4Z4 macrosatellite repeat on 4q35, causing loss of silencing and aberrant expression of the D4Z4-embedded DUX4 gene leading to disease. By an unknown mechanism, D4Z4 copy-number correlates with FSHD phenotype. Here we show that the DUX4 proximal promoter (DUX4p) is sufficient to nucleate the enrichment of both constitutive and facultative heterochromatin components and to mediate a copy-number dependent gene silencing. We found that both the CpG/GC dense DNA content and the repetitive nature of DUX4p arrays are important for their repressive ability. We showed that DUX4p mediates a copy number-dependent Polycomb Repressive Complex 1 (PRC1) recruitment, which is responsible for the copy-number dependent gene repression. Overall, we directly link genetic and epigenetic defects in FSHD by proposing a novel molecular explanation for the copy number-dependency in FSHD pathogenesis, and offer insight into the molecular functions of repeats in chromatin regulation.


Subject(s)
DNA Copy Number Variations , Homeodomain Proteins , Microsatellite Repeats , Muscular Dystrophy, Facioscapulohumeral , Polycomb Repressive Complex 1 , Cell Line , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism
3.
PLoS One ; 8(6): e67762, 2013.
Article in English | MEDLINE | ID: mdl-23840772

ABSTRACT

Quiescent satellite cells are myogenic progenitors that enable regeneration of skeletal muscle. One of the early events of satellite cell activation following myotrauma is the induction of the myogenic regulatory factor MyoD, which eventually induces terminal differentiation and muscle function gene expression. The purpose of this study was to elucidate the mechanism by which MyoD is induced during activation of satellite cells in mouse muscle undergoing regeneration. We show that Six1, a transcription factor essential for embryonic myogenesis, also regulates MyoD expression in muscle progenitor cells. Six1 knock-down by RNA interference leads to decreased expression of MyoD in myoblasts. Chromatin immunoprecipitation assays reveal that Six1 binds the Core Enhancer Region of MyoD. Further, transcriptional reporter assays demonstrate that Core Enhancer Region reporter gene activity in myoblasts and in regenerating muscle depends on the expression of Six1 and on Six1 binding sites. Finally, we provide evidence indicating that Six1 is required for the proper chromatin structure at the Core Enhancer Region, as well as for MyoD binding at its own enhancer. Together, our results reveal that MyoD expression in satellite cells depends on Six1, supporting the idea that Six1 plays an important role in adult myogenesis, in addition to its role in embryonic muscle formation.


Subject(s)
Homeodomain Proteins/genetics , Muscle, Skeletal/physiology , MyoD Protein/genetics , Satellite Cells, Skeletal Muscle/physiology , Stem Cells/physiology , Animals , Binding Sites/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Chromatin/genetics , Female , Gene Expression Regulation, Developmental/genetics , Genes, Reporter/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Muscle Development/genetics , Muscle Development/physiology , Muscle, Skeletal/metabolism , MyoD Protein/metabolism , Myoblasts/metabolism , Myoblasts/physiology , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Regeneration/genetics , Regeneration/physiology , Satellite Cells, Skeletal Muscle/metabolism , Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Genes Dev ; 27(11): 1247-59, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23723416

ABSTRACT

Alternate splicing contributes extensively to cellular complexity by generating protein isoforms with divergent functions. However, the role of alternate isoforms in development remains poorly understood. Mef2 transcription factors are essential transducers of cell signaling that modulate differentiation of many cell types. Among Mef2 family members, Mef2D is unique, as it undergoes tissue-specific splicing to generate a muscle-specific isoform. Since the ubiquitously expressed (Mef2Dα1) and muscle-specific (Mef2Dα2) isoforms of Mef2D are both expressed in muscle, we examined the relative contribution of each Mef2D isoform to differentiation. Using both in vitro and in vivo models, we demonstrate that Mef2D isoforms act antagonistically to modulate differentiation. While chromatin immunoprecipitation (ChIP) sequencing analysis shows that the Mef2D isoforms bind an overlapping set of genes, only Mef2Dα2 activates late muscle transcription. Mechanistically, the differential ability of Mef2D isoforms to activate transcription depends on their susceptibility to phosphorylation by protein kinase A (PKA). Phosphorylation of Mef2Dα1 by PKA provokes its association with corepressors. Conversely, exon switching allows Mef2Dα2 to escape this inhibitory phosphorylation, permitting recruitment of Ash2L for transactivation of muscle genes. Thus, our results reveal a novel mechanism in which a tissue-specific alternate splicing event has evolved that permits a ubiquitously expressed transcription factor to escape inhibitory signaling for temporal regulation of gene expression.


Subject(s)
Alternative Splicing , Cell Differentiation/genetics , Muscles/cytology , Muscles/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Animals , Chromatin Immunoprecipitation , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , Exons/genetics , Gene Expression Regulation/genetics , Genome/genetics , MEF2 Transcription Factors , Mice , Muscles/enzymology , Mutation/genetics , Myogenic Regulatory Factors/chemistry , Nuclear Proteins/metabolism , Organ Specificity/genetics , Phosphorylation/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics
5.
Stem Cell Rev Rep ; 8(2): 609-22, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22278133

ABSTRACT

Satellite cells represent the primary population of stem cells resident in skeletal muscle. These adult muscle stem cells facilitate the postnatal growth, remodeling, and regeneration of skeletal muscle. Given the remarkable regenerative potential of satellite cells, there is great promise for treatment of muscle pathologies such as the muscular dystrophies with this cell population. Various protocols have been developed which allow for isolation, enrichment, and expansion of satellite cell derived muscle stem cells. However, isolated satellite cells have yet to translate into effective modalities for therapeutic intervention. Broadening our understanding of satellite cells and their niche requirements should improve our in vivo and ex vivo manipulation of these cells to expedite their use for regeneration of diseased muscle. This review explores the fates of satellite cells as determined by their molecular signatures, ontogeny, and niche dependent programming.


Subject(s)
Cell Lineage , Satellite Cells, Skeletal Muscle/cytology , Body Patterning , Humans , Regenerative Medicine , Satellite Cells, Skeletal Muscle/metabolism , Signal Transduction , Stem Cell Niche
6.
Epigenetics ; 5(8): 691-5, 2010.
Article in English | MEDLINE | ID: mdl-20716948

ABSTRACT

MyoD is a master regulator of the skeletal muscle gene expression program. ChIP-Seq analysis has recently revealed that MyoD binds to a large number of genomic loci in differentiating myoblasts, yet only activates transcription at a subset of these genes. Here we discuss recent data suggesting that the ability of MyoD to mediate gene expression is regulated through the function of Polycomb and Trithorax Group proteins. Based on studies of the muscle-specific myog gene, we propose a model where the transcriptional activators Mef2d and Six4 mediate recruitment of Trithorax Group proteins Ash2L/MLL2 and UTX to MyoD-bound promoters to overcome the Polycomb-mediated repression of muscle genes. Modulation of the interaction between Ash2L/MLL2 and Mef2d by the p38α MAPK signaling pathway in turns provides fine-tuning of the muscle-specific gene expression program. Thus Mef2d, Six4, and p38α MAPK function coordinately as regulators of a master regulator to mediate expression of MyoD target genes.


Subject(s)
Gene Expression Regulation/physiology , Models, Biological , Muscle, Skeletal/metabolism , MyoD Protein/biosynthesis , Animals , Cell Differentiation/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genetic Loci/physiology , Histone Demethylases , Histone-Lysine N-Methyltransferase , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , MADS Domain Proteins/genetics , MADS Domain Proteins/metabolism , MAP Kinase Signaling System/physiology , MEF2 Transcription Factors , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , MyoD Protein/genetics , Myoblasts/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Myogenin/genetics , Myogenin/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organ Specificity/physiology , Polycomb-Group Proteins , Promoter Regions, Genetic/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
7.
EMBO J ; 29(8): 1401-11, 2010 Apr 21.
Article in English | MEDLINE | ID: mdl-20300060

ABSTRACT

Polycomb (PcG) and Trithorax (TrxG) group proteins act antagonistically to establish tissue-specific patterns of gene expression. The PcG protein Ezh2 facilitates repression by catalysing histone H3-Lys27 trimethylation (H3K27me3). For expression, H3K27me3 marks are removed and replaced by TrxG protein catalysed histone H3-Lys4 trimethylation (H3K4me3). Although H3K27 demethylases have been identified, the mechanism by which these enzymes are targeted to specific genomic regions to remove H3K27me3 marks has not been established. Here, we demonstrate a two-step mechanism for UTX-mediated demethylation at muscle-specific genes during myogenesis. Although the transactivator Six4 initially recruits UTX to the regulatory region of muscle genes, the resulting loss of H3K27me3 marks is limited to the region upstream of the transcriptional start site. Removal of the repressive H3K27me3 mark within the coding region then requires RNA Polymerase II (Pol II) elongation. Interestingly, blocking Pol II elongation on transcribed genes leads to increased H3K27me3 within the coding region, and formation of bivalent (H3K27me3/H3K4me3) chromatin domains. Thus, removal of repressive H3K27me3 marks by UTX occurs through targeted recruitment followed by spreading across the gene.


Subject(s)
Histones/metabolism , Muscle Development , Nuclear Proteins/metabolism , Animals , Cell Line , Creatine Kinase/metabolism , Genes , Histone Demethylases/metabolism , Homeodomain Proteins/metabolism , Methylation , Mice , Myoblasts/cytology , Myoblasts/metabolism , Myogenin/genetics , RNA Polymerase II/antagonists & inhibitors , RNA Polymerase II/metabolism , Trans-Activators/metabolism
8.
Dev Biol ; 332(1): 116-30, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19464283

ABSTRACT

Menin plays an established role in the differentiation of mesenchymal cells to the osteogenic lineage. Conversely, whether Menin influences the commitment of mesenschymal cells to the myogenic lineage, despite expression in the developing somite was previously unclear. We observed that Menin is down-regulated in C2C12 and C3H10T1/2 mesenchymal cells when muscle differentiation is induced. Moreover, maintenance of Menin expression by constitutive ectopic expression inhibited muscle cell differentiation. Reduction of Menin expression by siRNA technology results in precocious muscle differentiation and concomitantly attenuates BMP-2 induced osteogenesis. Reduced Menin expression antagonizes BMP-2 and TGF-beta1 mediated inhibition of myogenesis. Furthermore, Menin was found to directly interact with and potentiate the transactivation properties of Smad3 in response to TGF-beta1. Finally in concert with these observations, tissue-specific inactivation of Men1 in Pax3-expressing somite precursor cells leads to a patterning defect of rib formation and increased muscle mass in the intercostal region. These data invoke a pivotal role for Menin in the competence of mesenchymal cells to respond to TGF-beta1 and BMP-2 signals. Thus, by modulating cytokine responsiveness Menin functions to alter the balance of multipotent mesenchymal cell commitment to the osteogenic or myogenic lineages.


Subject(s)
Cell Lineage , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Muscle Development/genetics , Osteogenesis/genetics , Proto-Oncogene Proteins/metabolism , Animals , Bone Morphogenetic Protein 2/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Lineage/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Deletion , Humans , Intercostal Muscles/anatomy & histology , Intercostal Muscles/drug effects , Mesenchymal Stem Cells/drug effects , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Muscle Development/drug effects , MyoD Protein/metabolism , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Organ Size/drug effects , Organ Specificity/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Protein Binding/drug effects , Proto-Oncogene Proteins/genetics , Smad3 Protein/metabolism , Somites/cytology , Somites/drug effects , Somites/metabolism , Transforming Growth Factor beta1/metabolism
9.
J Biol Chem ; 284(29): 19679-93, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19439412

ABSTRACT

Skeletal myogenesis is potently regulated by the extracellular milieu of growth factors and cytokines. We observed that cardiotrophin-1 (CT-1), a member of the interleukin-6 (IL-6) family of cytokines, is a potent regulator of skeletal muscle differentiation. The normal up-regulation of myogenic marker genes, myosin heavy chain (MyHC), myogenic regulatory factors (MRFs), and myocyte enhancer factor 2s (MEF2s) were inhibited by CT-1 treatment. CT-1 also represses myogenin (MyoG) promoter activation. CT-1 activated two signaling pathways: signal transducer and activator of transcription 3 (STAT3), and mitogen-activated protein kinase kinase (MEK), a component of the extracellular signal-regulated MAPK (ERK) pathway. In view of the known connection between CT-1 and STAT3 activation, we surprisingly found that pharmacological blockade of STAT3 activity had no effect on the inhibition of myogenesis by CT-1 suggesting that STAT3 signaling is dispensable for myogenic repression. Conversely, MEK inhibition potently reversed the inhibition of myotube formation and attenuated the repression of MRF transcriptional activity mediated by CT-1. Taken together, these data indicate that CT-1 represses skeletal myogenesis through interference with MRF activity by activation of MEK/ERK signaling. In agreement with these in vitro observations, exogenous systemic expression of CT-1 mediated by adenoviral vector delivery increased the number of myonuclei in normal post-natal mouse skeletal muscle and also delayed skeletal muscle regeneration induced by cardiotoxin injection. The expression pattern of CT-1 in embryonic and post-natal skeletal muscle and in vivo effects of CT-1 on myogenesis implicate CT-1 in the maintenance of the undifferentiated state in muscle progenitor cells.


Subject(s)
Cell Differentiation/drug effects , Cytokines/pharmacology , Myoblasts, Skeletal/drug effects , Recombinant Proteins/pharmacology , Animals , Blotting, Western , Butadienes/pharmacology , Cell Line , Cytokines/genetics , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoprecipitation , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , Mice , Muscle Development/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Nitriles/pharmacology , Phosphorylation/drug effects , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Transfection
10.
Mol Cell Biol ; 28(9): 2952-70, 2008 May.
Article in English | MEDLINE | ID: mdl-18299387

ABSTRACT

Activation of protein kinase A (PKA) by elevation of the intracellular cyclic AMP (cAMP) level inhibits skeletal myogenesis. Previously, an indirect modulation of the myogenic regulatory factors (MRFs) was implicated as the mechanism. Because myocyte enhancer factor 2 (MEF2) proteins are key regulators of myogenesis and obligatory partners for the MRFs, here we assessed whether these proteins could be involved in PKA-mediated myogenic repression. Initially, in silico analysis revealed several consensus PKA phosphoacceptor sites on MEF2, and subsequent analysis by in vitro kinase assays indicated that PKA directly and efficiently phosphorylates MEF2D. Using mass spectrometric determination of phosphorylated residues, we document that MEF2D serine 121 and serine 190 are targeted by PKA. Transcriptional reporter gene assays to assess MEF2D function revealed that PKA potently represses the transactivation properties of MEF2D. Furthermore, engineered mutation of MEF2D PKA phosphoacceptor sites (serines 121 and 190 to alanine) rendered a PKA-resistant MEF2D protein, which efficiently rescues myogenesis from PKA-mediated repression. Concomitantly, increased intracellular cAMP-mediated PKA activation also resulted in an enhanced nuclear accumulation of histone deacetylase 4 (HDAC4) and a subsequent increase in the MEF2D-HDAC4 repressor complex. Collectively, these data identify MEF2D as a primary target of PKA signaling in myoblasts that leads to inhibition of the skeletal muscle differentiation program.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Muscle Cells/cytology , Muscle Development/physiology , Muscle, Skeletal/physiology , Myogenic Regulatory Factors/physiology , Amino Acid Sequence , Animals , Cell Differentiation , Cells, Cultured , Cyclic AMP/metabolism , Histone Deacetylases/physiology , MEF2 Transcription Factors , Mice , Molecular Sequence Data , Muscle Cells/physiology , Muscle, Skeletal/cytology , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
11.
Mol Cell Biol ; 26(16): 6248-60, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16880533

ABSTRACT

Transforming growth factor beta1 (TGF-beta1) and myostatin signaling, mediated by the same Smad downstream effectors, potently repress skeletal muscle cell differentiation. Smad7 inhibits these cytokine signaling pathways. The role of Smad7 during skeletal muscle cell differentiation was assessed. In these studies, we document that increased expression of Smad7 abrogates myostatin- but not TGF-beta1-mediated repression of myogenesis. Further, constitutive expression of exogenous Smad7 potently enhanced skeletal muscle differentiation and cellular hypertrophy. Conversely, targeting of endogenous Smad7 by small interfering RNA inhibited C2C12 muscle cell differentiation, indicating an essential role for Smad7 during myogenesis. Congruent with a role for Smad7 in myogenesis, we observed that the muscle regulatory factor (MyoD) binds to and transactivates the Smad7 proximal promoter region. Finally, we document that Smad7 directly interacts with MyoD and enhances MyoD transcriptional activity. Thus, Smad7 cooperates with MyoD, creating a positive loop to induce Smad7 expression and to promote MyoD driven myogenesis. Taken together, these data implicate Smad7 as a fundamental regulator of differentiation in skeletal muscle cells.


Subject(s)
Cell Differentiation , Muscle, Skeletal/cytology , Smad7 Protein/metabolism , Activin Receptors, Type II/metabolism , Animals , Cells, Cultured , Fibroblasts/cytology , Genes, Dominant , Mice , Models, Biological , Muscle Development , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Mutation/genetics , MyoD Protein/metabolism , Myoblasts/cytology , Myostatin , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Small Interfering/genetics , Transforming Growth Factor beta/metabolism
12.
Gastrointest Endosc ; 62(2): 245-50, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16046988

ABSTRACT

BACKGROUND: Pancreatitis is the most common major complication of ERCP. Efforts have been made to identify pharmacologic agents capable of reducing its incidence and severity. The aim of this trial was to determine whether prophylactic allopurinol, an inhibitor of oxygen-derived free radical production, would reduce the frequency and severity of post-ERCP pancreatitis. Methods A total of 701 patients were randomized to receive either allopurinol or placebo 4 hours and 1 hour before ERCP. A database was prospectively collected by a defined protocol on patients who underwent ERCP. Standardized criteria were used to diagnose and grade the severity of postprocedure pancreatitis. RESULTS: The groups were similar with regard to patient demographics and to patient and procedure risk factors for pancreatitis. The overall incidence of pancreatitis was 12.55%. It occurred in 46 of 355 patients in the allopurinol group (12.96%) and in 42 of 346 patients in the control group (12.14%; p = 0.52). The pancreatitis was graded mild in 7.89%, moderate in 4.51%, and severe in 0.56% of the allopurinol group, and mild in 6.94%, moderate in 4.62%, and severe in 0.58% of the control group. There was no significant difference between the groups in the frequency or the severity of pancreatitis. CONCLUSIONS: Prophylactic oral allopurinol did not reduce the frequency or the severity of post-ERCP pancreatitis.


Subject(s)
Allopurinol/administration & dosage , Cholangiopancreatography, Endoscopic Retrograde/adverse effects , Free Radical Scavengers/administration & dosage , Pancreatitis/prevention & control , Administration, Oral , Double-Blind Method , Female , Humans , Male , Middle Aged , Pancreatitis/etiology , Prospective Studies , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...