Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
PLoS Genet ; 9(4): e1003445, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23637619

RESUMO

We report the identification and characterization of a previously unknown suppressor of myopathy caused by expansion of CUG repeats, the mutation that triggers Myotonic Dystrophy Type 1 (DM1). We screened a collection of genes encoding RNA-binding proteins as candidates to modify DM1 pathogenesis using a well established Drosophila model of the disease. The screen revealed smaug as a powerful modulator of CUG-induced toxicity. Increasing smaug levels prevents muscle wasting and restores muscle function, while reducing its function exacerbates CUG-induced phenotypes. Using human myoblasts, we show physical interactions between human Smaug (SMAUG1/SMAD4A) and CUGBP1. Increased levels of SMAUG1 correct the abnormally high nuclear accumulation of CUGBP1 in myoblasts from DM1 patients. In addition, augmenting SMAUG1 levels leads to a reduction of inactive CUGBP1-eIF2α translational complexes and to a correction of translation of MRG15, a downstream target of CUGBP1. Therefore, Smaug suppresses CUG-mediated muscle wasting at least in part via restoration of translational activity of CUGBP1.


Assuntos
Distrofia Miotônica , Proteínas de Ligação a RNA , Regulação da Expressão Gênica , Humanos , Mioblastos/metabolismo , Distrofia Miotônica/genética , Proteínas de Ligação a RNA/genética
2.
Rev Neurosci ; 21(1): 19-28, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20458885

RESUMO

Expansion of non-coding CTG and CCTG repeats in the 3' UTR of the myotonin protein kinase (DMPK) gene in Myotonic Dystrophy type 1 (DM1) and in the intron 1 of Zinc Finger Protein 9 (ZNF9) in Myotonic Dystrophy type 2 (DM2) represent typical non-coding mutations that cause the diseases mainly through transdominant effect on the RNA metabolism (splicing, translation and RNA stability). The commonly recognized RNA gain-of-function mechanism of DM1 and DM2 suggests that the mutant CUG and CCUG RNAs play a critical role in myotonic dystrophies (DMs) without a significant role of DMPK and ZNF9. Recent studies have shown that the molecular pathogenesis of DM2 also involves the protein product of the ZNF9 gene. CCUG repeats reduce ZNF9 protein, a translational regulator of the terminal oligo-pyrimidine tract (TOP) mRNAs encoding proteins of translational apparatus. Thus, in DM2 cells, expansion of CCUG repeats affects not only multiple RNAs, but also down-regulates ZNF9 which in turn reduces translation of the TOP-containing mRNAs and diminishes the rate of global protein synthesis. In this review, we discuss the role of expansion of CCUG repeats in the reduction of ZNF9-mediated regulation of the rate of protein synthesis in DM2 and the consequences of this reduction in the multi-systemic phenotype of DM2.


Assuntos
Distrofia Miotônica/genética , Distrofia Miotônica/metabolismo , Biossíntese de Proteínas/fisiologia , Proteínas de Ligação a RNA/metabolismo , Sequências de Repetição em Tandem/genética , Animais , Humanos , Proteínas de Ligação a RNA/genética
3.
Am J Pathol ; 175(2): 748-62, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19590039

RESUMO

Myotonic dystrophy 2 (DM2) is a multisystem skeletal muscle disease caused by an expansion of tetranucleotide CCTG repeats, the transcription of which results in the accumulation of untranslated CCUG RNA. In this study, we report that CCUG repeats both bind to and misregulate the biological functions of cytoplasmic multiprotein complexes. Two CCUG-interacting complexes were subsequently purified and analyzed. A major component of one of the complexes was found to be the 20S catalytic core complex of the proteasome. The second complex was found to contain CUG triplet repeat RNA-binding protein 1 (CUGBP1) and the translation initiation factor eIF2. Consistent with the biological functions of the 20S proteasome and the CUGBP1-eIF2 complexes, the stability of short-lived proteins and the levels of the translational targets of CUGBP1 were shown to be elevated in DM2 myoblasts. We found that the overexpression of CCUG repeats in human myoblasts from unaffected patients, in C2C12 myoblasts, and in a DM2 mouse model alters protein translation and degradation, similar to the alterations observed in DM2 patients. Taken together, these findings show that RNA CCUG repeats misregulate protein turnover on both the levels of translation and proteasome-mediated protein degradation.


Assuntos
Repetições de Microssatélites , Distrofia Miotônica/metabolismo , Biossíntese de Proteínas , Proteínas/metabolismo , Regiões não Traduzidas/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Fator de Iniciação 2 em Eucariotos/metabolismo , Humanos , Camundongos , Mioblastos/metabolismo , Distrofia Miotônica/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Regiões não Traduzidas/genética
4.
Exp Cell Res ; 314(11-12): 2266-78, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18570922

RESUMO

Differentiation of myocytes is impaired in patients with myotonic dystrophy type 1, DM1. CUG repeat binding protein, CUGBP1, is a key regulator of translation of proteins that are involved in muscle development and differentiation. In this paper, we present evidence that RNA-binding activity of CUGBP1 and its interactions with initiation translation complex eIF2 are differentially regulated during myogenesis by specific phosphorylation and that this regulation is altered in DM1. In normal myoblasts, Akt kinase phosphorylates CUGBP1 at Ser28 and increases interactions of CUGBP1 with cyclin D1 mRNA. During differentiation, CUGBP1 is phosphorylated by cyclinD3-cdk4/6 at Ser302, which increases CUGBP1 binding with p21 and C/EBPbeta mRNAs. While cyclin D3 and cdk4 are elevated in normal myotubes; DM1 differentiating cells do not increase these proteins. In normal myotubes, CUGBP1 interacts with cyclin D3/cdk4/6 and eIF2; however, interactions of CUGBP1 with eIF2 are reduced in DM1 differentiating cells and correlate with impaired muscle differentiation in DM1. Ectopic expression of cyclin D3 in DM1 cells increases the CUGBP1-eIF2 complex, corrects expression of differentiation markers, myogenin and desmin, and enhances fusion of DM1 myoblasts. Thus, normalization of cyclin D3 might be a therapeutic approach to correct differentiation of skeletal muscle in DM1 patients.


Assuntos
Diferenciação Celular/fisiologia , Ciclinas/metabolismo , Desenvolvimento Muscular/fisiologia , Músculo Esquelético , Mioblastos/fisiologia , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proteínas CELF1 , Fusão Celular , Linhagem Celular , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina D3 , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Ciclinas/genética , Humanos , Camundongos , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Mioblastos/citologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas de Ligação a RNA/genética , Serina/metabolismo , Transdução de Sinais/fisiologia
5.
Mol Cell Biol ; 22(20): 7242-57, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12242300

RESUMO

We previously identified an RNA binding protein, CUGBP1, which binds to GCN repeats located within the 5' region of C/EBPbeta mRNAs and regulates translation of C/EBPbeta isoforms. To further investigate the role of RNA binding proteins in the posttranscriptional control of C/EBP proteins, we purified additional RNA binding proteins that interact with GC-rich RNAs and that may regulate RNA processing. In HeLa cells, the majority of GC-rich RNA binding proteins are associated with endogenous RNA transcripts. The separation of these proteins from endogenous RNA identified several proteins in addition to CUGBP1 that specifically interact with the GC-rich 5' region of C/EBPbeta mRNA. One of these proteins was purified to homogeneity and was identified as calreticulin (CRT). CRT is a multifunctional protein involved in several biological processes, including interaction with and regulation of rubella virus RNA processing. Our data demonstrate that both CUGBP1 and CRT interact with GCU repeats within myotonin protein kinase and with GCN repeats within C/EBPalpha and C/EBPbeta mRNAs. GCN repeats within these mRNAs form stable SL structures. The interaction of CRT with SL structures of C/EBPbeta and C/EBPalpha mRNAs leads to inhibition of translation of C/EBP proteins in vitro and in vivo. Deletions or mutations abolishing the formation of SL structures within C/EBPalpha and C/EBPbeta mRNAs lead to a failure of CRT to inhibit translation of C/EBP proteins. CRT-dependent inhibition of C/EBPalpha is sufficient to block the growth-inhibitory activity of C/EBPalpha. This finding further defines the molecular mechanism for posttranscriptional regulation of the C/EBPalpha and C/EBPbeta proteins.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteínas de Ligação ao Cálcio/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo , Ribonucleoproteínas/metabolismo , Sítios de Ligação , Calreticulina , Divisão Celular , Sistema Livre de Células , Células HeLa , Humanos
6.
Cell Death Dis ; 7(10): e2426, 2016 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-27763639

RESUMO

Myoblasts are mononucleated precursors of myofibers; they persist in mature skeletal muscles for growth and regeneration post injury. During myotonic dystrophy type 1 (DM1), a complex autosomal-dominant neuromuscular disease, the differentiation of skeletal myoblasts into functional myotubes is impaired, resulting in muscle wasting and weakness. The mechanisms leading to this altered differentiation are not fully understood. Here, we demonstrate that the calcium- and voltage-dependent potassium channel, KCa1.1 (BK, Slo1, KCNMA1), regulates myoblast proliferation, migration, and fusion. We also show a loss of plasma membrane expression of the pore-forming α subunit of KCa1.1 in DM1 myoblasts. Inhibiting the function of KCa1.1 in healthy myoblasts induced an increase in cytosolic calcium levels and altered nuclear factor kappa B (NFκB) levels without affecting cell survival. In these normal cells, KCa1.1 block resulted in enhanced proliferation and decreased matrix metalloproteinase secretion, migration, and myotube fusion, phenotypes all observed in DM1 myoblasts and associated with disease pathogenesis. In contrast, introducing functional KCa1.1 α-subunits into DM1 myoblasts normalized their proliferation and rescued expression of the late myogenic marker Mef2. Our results identify KCa1.1 channels as crucial regulators of skeletal myogenesis and suggest these channels as novel therapeutic targets in DM1.


Assuntos
Diferenciação Celular , Movimento Celular , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Cálcio/metabolismo , Fusão Celular , Membrana Celular/metabolismo , Proliferação de Células , Células Cultivadas , Humanos , Espaço Intracelular/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Distrofia Miotônica/patologia , NF-kappa B/metabolismo
7.
Neuromuscul Disord ; 24(3): 227-40, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24332166

RESUMO

The prevailing pathomechanistic paradigm for myotonic dystrophy (DM) is that aberrant expression of embryonic/fetal mRNA/protein isoforms accounts for most aspects of the pleiotropic phenotype. To identify aberrant isoforms in skeletal muscle of DM1 and DM2 patients, we performed exon-array profiling and RT-PCR validation on the largest DM sample set to date, including Duchenne, Becker and tibial muscular dystrophy (NMD) patients as disease controls, and non-disease controls. Strikingly, most expression and splicing changes in DM patients were shared with NMD controls. Comparison between DM and NMD identified almost no significant differences. We conclude that DM1 and DM2 are essentially identical for dysregulation of gene expression, and DM expression changes represent a subset of broader spectrum dystrophic changes. We found no evidence for qualitative splicing differences between DM1 and DM2. While some DM-specific splicing differences exist, most of the DM splicing differences were also seen in NMD controls. SSBP3 exon 6 missplicing was observed in all diseased muscle and led to reduced protein. We conclude there is no widespread DM-specific spliceopathy in skeletal muscle and suggest that missplicing in DM (and NMD) may not be the driving mechanism for the muscle pathology, since the same pathways show expression changes unrelated to splicing.


Assuntos
Expressão Gênica , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Transtornos Miotônicos/genética , Distrofia Miotônica/genética , Splicing de RNA , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Éxons , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Distrofias Musculares/metabolismo , Transtornos Miotônicos/metabolismo , Distrofia Miotônica/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Adulto Jovem
8.
Histol Histopathol ; 28(9): 1089-98, 2013 09.
Artigo em Inglês | MEDLINE | ID: mdl-23536431

RESUMO

Neuromuscular diseases Myotonic Dystrophies type 1 and type 2 (DM1 and DM2) are caused by unstable CTG and CCTG repeat expansions and have highly complex molecular mechanisms. DM1 is caused by the expansion of CTG repeats in the 3' UTR of the gene coding for Dystrophia Myotonica-Protein Kinase (DMPK). In DM2, intronic CCTG repeats are located in a gene encoding the Zinc Finger Protein 9 (ZNF9, also known as Cellular Nucleic Acid Binding Protein, CNBP). Both expansions cause pathologies through RNA CUG and CCUG repeats, which have toxic effects on the processing of many RNAs in the patients' tissues. The pathogenic role of CUG and CCUG repeats in the mis-regulation of alternative splicing, mediated by RNA-binding proteins CUGBP1 and MBNL1, has been discussed in a number of excellent reviews. Recent reports suggest that mutant RNA repeats affect several other RNA-binding proteins such as Staufen1 and the DEAD-box RNA helicase p68 (DDX5). Since CUGBP1, Staufen1 and p68 have many functions in cytoplasm, including regulation of protein translation, it is predicted that the alterations of these proteins in DM cells might have a toxic effect on global protein turnover. In this mini-review, we will summarize observations showing the role of RNA-binding proteins, CUGBP1 and ZNF9, in protein turnover in DM1 and in DM2. We will also discuss a possible role of misbalanced protein turnover in the age-dependent progression of DM1 and in a late onset of DM2.


Assuntos
Regulação da Expressão Gênica , Distrofia Miotônica/patologia , Proteínas/química , Envelhecimento , Animais , Proteínas CELF1 , Citoplasma/metabolismo , Proteínas do Citoesqueleto/metabolismo , RNA Helicases DEAD-box/metabolismo , Progressão da Doença , Homeostase , Humanos , Distrofia Miotônica/metabolismo , Proteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo
9.
J Clin Invest ; 122(12): 4461-72, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23160194

RESUMO

Myotonic dystrophy type 1 (DM1) is a complex neuromuscular disease characterized by skeletal muscle wasting, weakness, and myotonia. DM1 is caused by the accumulation of CUG repeats, which alter the biological activities of RNA-binding proteins, including CUG-binding protein 1 (CUGBP1). CUGBP1 is an important skeletal muscle translational regulator that is activated by cyclin D3-dependent kinase 4 (CDK4). Here we show that mutant CUG repeats suppress Cdk4 signaling by increasing the stability and activity of glycogen synthase kinase 3ß (GSK3ß). Using a mouse model of DM1 (HSA(LR)), we found that CUG repeats in the 3' untranslated region (UTR) of human skeletal actin increase active GSK3ß in skeletal muscle of mice, prior to the development of skeletal muscle weakness. Inhibition of GSK3ß in both DM1 cell culture and mouse models corrected cyclin D3 levels and reduced muscle weakness and myotonia in DM1 mice. Our data predict that compounds normalizing GSK3ß activity might be beneficial for improvement of muscle function in patients with DM1.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Músculo Esquelético/enzimologia , Distrofia Miotônica/enzimologia , Adulto , Animais , Células CHO , Cricetinae , Ciclina D3/metabolismo , Estabilidade Enzimática , Feminino , Regulação Enzimológica da Expressão Gênica , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Lítio/farmacologia , Lítio/uso terapêutico , Masculino , Camundongos , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/patologia , Força Muscular/efeitos dos fármacos , Músculo Esquelético/patologia , Distrofia Miotônica/tratamento farmacológico , Distrofia Miotônica/patologia , Fosforilação , Processamento de Proteína Pós-Traducional , Tiadiazóis/farmacologia , Tiadiazóis/uso terapêutico
10.
J Vis Exp ; (45)2010 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-21085106

RESUMO

Glucose is the main source of energy for the body, requiring constant regulation of its blood concentration. Insulin release by the pancreas induces glucose uptake by insulin-sensitive tissues, most notably the brain, skeletal muscle, and adipocytes. Patients suffering from type-2 diabetes and/or obesity often develop insulin resistance and are unable to control their glucose homeostasis. New insights into the mechanisms of insulin resistance may provide new treatment strategies for type-2 diabetes. The GLUT family of glucose transporters consists of thirteen members distributed on different tissues throughout the body. Glucose transporter type 4 (GLUT4) is the major transporter that mediates glucose uptake by insulin sensitive tissues, such as the skeletal muscle. Upon binding of insulin to its receptor, vesicles containing GLUT4 translocate from the cytoplasm to the plasma membrane, inducing glucose uptake. Reduced GLUT4 translocation is one of the causes of insulin resistance in type-2 diabetes. The translocation of GLUT4 from the cytoplasm to the plasma membrane can be visualized by immunocytochemistry, using fluorophore-conjugated GLUT4-specific antibodies. Here, we describe a technique to quantify total amounts of GLUT4 translocation to the plasma membrane of cells during a chosen duration, using flow cytometry. This protocol is rapid (less than 4 hours, including incubation with insulin) and allows the analysis of as few as 3,000 cells or as many as 1 million cells per condition in a single experiment. It relies on anti-GLUT4 antibodies directed to an external epitope of the transporter that bind to it as soon as it is exposed to the extracellular medium after translocation to the plasma membrane.


Assuntos
Citometria de Fluxo/métodos , Transportador de Glucose Tipo 4/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Transportador de Glucose Tipo 4/análise , Humanos
12.
J Biol Chem ; 281(43): 32806-19, 2006 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-16931514

RESUMO

The RNA-binding protein CUGBP1 regulates translation of proteins in a variety of biological processes. In this study, we show that aging liver increases CUGBP1 translational activities by induction of a high molecular weight protein-protein complex of CUGBP1. The complex contains CUGBP1, subunits alpha, beta, and gamma of the initiation translation factor eIF2, and four proteins of the endoplasmic reticulum, eR90, CRT, eR60, and Grp78. The induction of the CUGBP1-eIF2 complex in old livers is associated with the elevation of protein levels of CUGBP1 and with the hyper-phosphorylation of CUGBP1 by a cyclin D3-cdk4 kinase, activity of which is increased with age. We have examined the role of the elevation of CUGBP1 and the role of cyclin D3-cdk4-mediated phosphorylation of CUGBP1 in the formation of the CUGBP1-eIF2 complex by using CUGBP1 transgenic mice and young animals expressing high levels of cyclin D3 after injection with cyclin D3 plasmid. These studies showed that both the increased levels of CUGBP1 and cdk4-mediated hyper-phosphorylation of CUGBP1 are involved in the age-associated induction of the CUGBP1-eIF2 complex. The CUGBP1-eIF2 complex is bound to C/EBPbeta mRNA in the liver of old animals, and this binding correlates with the increased amounts of liver-enriched activator protein and liver-enriched inhibitory protein. Consistent with these observations, the purified CUGBP1-eIF2 complex binds to the 5' region of C/EBPbeta mRNA and significantly increases translation of the three isoforms of C/EBPbeta in a cell-free translation system, in cultured cells, and in the liver. Thus, these studies demonstrated that age-mediated induction of the CUGBP1-eIF2 complex changes translation of C/EBPbeta in old livers.


Assuntos
Envelhecimento/fisiologia , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Fator de Iniciação 2 em Eucariotos/metabolismo , Fígado/metabolismo , Biossíntese de Proteínas , Proteínas de Ligação a RNA/metabolismo , Animais , Proteínas CELF1 , Carcinoma Hepatocelular/patologia , Linhagem Celular , Linhagem Celular Tumoral , Chaperona BiP do Retículo Endoplasmático , Proteínas de Fluorescência Verde/metabolismo , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Transgênicos , RNA Mensageiro/metabolismo , Transfecção
13.
J Biol Chem ; 280(21): 20549-57, 2005 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-15788409

RESUMO

Expression of a dominant negative 20-kDa isoform of CCAAT/enhancer-binding protein (C/EBPbeta), LIP, is increased in proliferating livers and in tumor cells. Two RNA-binding proteins, CUGBP1 and calreticulin, have been implicated in the translational regulation of C/EBPbeta. In this paper, we present evidence showing several critical steps by which liver increases translation of LIP after partial hepatectomy. At early stages after partial hepatectomy, liver activates CUGBP1 by a hyperphosphorylation. The activated CUGBP1 binds to the 5' region of C/EBPbeta mRNA and replaces calreticulin, which partially represses translation of C/EBPbeta in quiescent livers. The hyperphosphorylated CUGBP1 also interacts with the alpha and beta subunits of initiation factor eIF2. Our data demonstrate that the interaction of CUGBP1 with the eIF2alpha enhances the association of CUGBP1 with ribosomes and correlates with increased translation of LIP in the liver after partial hepatectomy. Our data support the hypothesis that CUGBP1 increases translation of LIP by the interaction with the eIF2alpha subunit. This facilitates subsequent recruitment of larger numbers of ribosomes to initiate translation of LIP.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/genética , Fator de Iniciação 2 em Eucariotos/metabolismo , Regulação da Expressão Gênica , Proteínas de Ligação a RNA/fisiologia , Animais , Western Blotting , Proteínas CELF1 , Calreticulina/metabolismo , Proteínas de Fluorescência Verde/genética , Células HeLa , Hepatectomia , Humanos , Fígado/metabolismo , Camundongos , Fosforilação , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas Recombinantes de Fusão , Ribossomos/metabolismo , Transfecção
14.
J Biol Chem ; 279(13): 13129-39, 2004 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-14722059

RESUMO

Accumulation of RNA CUG repeats in myotonic dystrophy type 1 (DM1) patients leads to the induction of a CUG-binding protein, CUGBP1, which increases translation of several proteins that are required for myogenesis. In this paper, we examine the role of overexpression of CUGBP1 in DM1 muscle pathology using transgenic mice that overexpress CUGBP1 in skeletal muscle. Our data demonstrate that the elevation of CUGBP1 in skeletal muscle causes overexpression of MEF2A and p21 to levels that are significantly higher than those in skeletal muscle of wild type animals. A similar induction of these proteins is observed in skeletal muscle of DM1 patients with increased levels of CUGBP1. Immunohistological analysis showed that the skeletal muscle from mice overexpressing CUGBP1 is characterized by a developmental delay, muscular dystrophy, and myofiber-type switch: increase of slow/oxidative fibers and the reduction of fast fibers. Examination of molecular mechanisms by which CUGBP1 up-regulates MEF2A shows that CUGBP1 increases translation of MEF2A via direct interaction with GCN repeats located within MEF2A mRNA. Our data suggest that CUGBP1-mediated overexpression of MEF2A and p21 inhibits myogenesis and contributes to the development of muscle deficiency in DM1 patients.


Assuntos
Músculos/metabolismo , Proteínas de Ligação a RNA/biossíntese , Animais , Northern Blotting , Western Blotting , Peso Corporal , Proteínas CELF1 , Sistema Livre de Células/metabolismo , Células Cultivadas , Reagentes de Ligações Cruzadas/farmacologia , Proteínas de Ligação a DNA/química , Fibroblastos/metabolismo , Genótipo , Humanos , Imuno-Histoquímica , Proteínas de Domínio MADS , Fatores de Transcrição MEF2 , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Músculo Esquelético/metabolismo , Músculos/citologia , Fatores de Regulação Miogênica , Distrofia Miotônica/metabolismo , Plasmídeos/metabolismo , Ligação Proteica , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , RNA/química , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/química , Fatores de Transcrição/química , Transgenes , Raios Ultravioleta , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA