Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 294(18): 7360-7376, 2019 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-30837270

RESUMO

Oculopharyngeal muscular dystrophy (OPMD) is a late-onset, primarily autosomal dominant disease caused by a short GCN expansion in the PABPN1 (polyadenylate-binding protein nuclear 1) gene that results in an alanine expansion at the N terminus of the PABPN1 protein. Expression of alanine-expanded PABPN1 is linked to the formation of nuclear aggregates in tissues from individuals with OPMD. However, as with other nuclear aggregate-associated diseases, controversy exists over whether these aggregates are the direct cause of pathology. An emerging hypothesis is that a loss of PABPN1 function and/or aberrant protein interactions contribute to pathology in OPMD. Here, we present the first global proteomic analysis of the protein interactions of WT and alanine-expanded PABPN1 in skeletal muscle tissue. These data provide both insight into the function of PABPN1 in muscle and evidence that the alanine expansion alters the protein-protein interactions of PABPN1. We extended this analysis to demonstrate altered complex formation with and loss of function of TDP-43 (TAR DNA-binding protein 43), which we show interacts with alanine-expanded but not WT PABPN1. The results from our study support a model where altered protein interactions with alanine-expanded PABPN1 that lead to loss or gain of function could contribute to pathology in OPMD.


Assuntos
Alanina/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular Oculofaríngea/metabolismo , Proteínas Nucleares/metabolismo , Proteína I de Ligação a Poli(A)/metabolismo , Proteômica , Animais , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Eletroporação , Feminino , Masculino , Camundongos , Peso Molecular , Distrofia Muscular Oculofaríngea/genética , Proteína I de Ligação a Poli(A)/genética , Estudo de Prova de Conceito , Ligação Proteica
2.
J Cell Sci ; 131(3)2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29361530

RESUMO

Skeletal muscle is primarily composed of large myofibers containing thousands of post-mitotic nuclei distributed throughout a common cytoplasm. Protein production and localization in specialized myofiber regions is crucial for muscle function. Myonuclei differ in transcriptional activity and protein accumulation, but how these differences among nuclei sharing a cytoplasm are achieved is unknown. Regulated nuclear import of proteins is one potential mechanism for regulating transcription spatially and temporally in individual myonuclei. The best-characterized nuclear localization signal (NLS) in proteins is the classical NLS (cNLS), but many other NLS motifs exist. We examined cNLS and non-cNLS reporter protein import using multinucleated muscle cells generated in vitro, revealing that cNLS and non-cNLS nuclear import differs among nuclei in the same cell. Investigation of cNLS nuclear import rates in isolated myofibers ex vivo confirmed differences in nuclear import rates among myonuclei. Analyzing nuclear import throughout myogenesis revealed that cNLS and non-cNLS import varies during differentiation. Taken together, our results suggest that both spatial and temporal regulation of nuclear import pathways are important in muscle cell differentiation and protein regionalization in myofibers.


Assuntos
Núcleo Celular/metabolismo , Células Musculares/citologia , Células Musculares/metabolismo , Músculo Esquelético/citologia , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Diferenciação Celular , Células Cultivadas , Masculino , Camundongos Endogâmicos C57BL , Desenvolvimento Muscular , Fibras Musculares Esqueléticas/metabolismo , Sinais de Localização Nuclear/metabolismo , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/metabolismo
3.
Nucleic Acids Res ; 46(15): 7643-7661, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-29939290

RESUMO

RNA processing is critical for proper spatial and temporal control of gene expression. The ubiquitous nuclear polyadenosine RNA binding protein, PABPN1, post-transcriptionally regulates multiple steps of gene expression. Mutations in the PABPN1 gene expanding an N-terminal alanine tract in the PABPN1 protein from 10 alanines to 11-18 alanines cause the muscle-specific disease oculopharyngeal muscular dystrophy (OPMD), which affects eyelid, pharynx, and proximal limb muscles. Previous work revealed that the Pabpn1 transcript is unstable, contributing to low steady-state Pabpn1 mRNA and protein levels in vivo, specifically in skeletal muscle, with even lower levels in muscles affected in OPMD. Thus, low levels of PABPN1 protein could predispose specific tissues to pathology in OPMD. However, no studies have defined the mechanisms that regulate Pabpn1 expression. Here, we define multiple cis-regulatory elements and a trans-acting factor, HuR, which regulate Pabpn1 expression specifically in mature muscle in vitro and in vivo. We exploit multiple models including C2C12 myotubes, primary muscle cells, and mice to determine that HuR decreases Pabpn1 expression. Overall, we have uncovered a mechanism in mature muscle that negatively regulates Pabpn1 expression in vitro and in vivo, which could provide insight to future studies investigating therapeutic strategies for OPMD treatment.


Assuntos
Proteína Semelhante a ELAV 1/genética , Regulação da Expressão Gênica , Proteína I de Ligação a Poli(A)/genética , Proteínas de Ligação a RNA/genética , Animais , Linhagem Celular , Modelos Animais de Doenças , Proteína Semelhante a ELAV 1/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/metabolismo , Distrofia Muscular Oculofaríngea/patologia , Mutação , Células NIH 3T3 , Proteína I de Ligação a Poli(A)/metabolismo , Proteínas de Ligação a RNA/metabolismo
4.
Hum Mol Genet ; 26(17): 3235-3252, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28575395

RESUMO

Oculopharyngeal muscular dystrophy (OPMD) is a late onset disease caused by polyalanine expansion in the poly(A) binding protein nuclear 1 (PABPN1). Several mouse models have been generated to study OPMD; however, most of these models have employed transgenic overexpression of alanine-expanded PABPN1. These models do not recapitulate the OPMD patient genotype and PABPN1 overexpression could confound molecular phenotypes. We have developed a knock-in mouse model of OPMD (Pabpn1+/A17) that contains one alanine-expanded Pabpn1 allele under the control of the native promoter and one wild-type Pabpn1 allele. This mouse is the closest available genocopy of OPMD patients. We show that Pabpn1+/A17 mice have a mild myopathic phenotype in adult and aged animals. We examined early molecular and biochemical phenotypes associated with expressing native levels of A17-PABPN1 and detected shorter poly(A) tails, modest changes in poly(A) signal (PAS) usage, and evidence of mitochondrial damage in these mice. Recent studies have suggested that a loss of PABPN1 function could contribute to muscle pathology in OPMD. To investigate a loss of function model of pathology, we generated a heterozygous Pabpn1 knock-out mouse model (Pabpn1+/Δ). Like the Pabpn1+/A17 mice, Pabpn1+/Δ mice have mild histologic defects, shorter poly(A) tails, and evidence of mitochondrial damage. However, the phenotypes detected in Pabpn1+/Δ mice only partially overlap with those detected in Pabpn1+/A17 mice. These results suggest that loss of PABPN1 function could contribute to but may not completely explain the pathology detected in Pabpn1+/A17 mice.


Assuntos
Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/metabolismo , Proteína I de Ligação a Poli(A)/genética , Proteína I de Ligação a Poli(A)/metabolismo , Animais , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Genótipo , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular Oculofaríngea/patologia , Peptídeos , Fenótipo
5.
Nucleic Acids Res ; 45(18): 10706-10725, 2017 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-28977530

RESUMO

The polyadenylate binding protein 1 (PABPN1) is a ubiquitously expressed RNA binding protein vital for multiple steps in RNA metabolism. Although PABPN1 plays a critical role in the regulation of RNA processing, mutation of the gene encoding this ubiquitously expressed RNA binding protein causes a specific form of muscular dystrophy termed oculopharyngeal muscular dystrophy (OPMD). Despite the tissue-specific pathology that occurs in this disease, only recently have studies of PABPN1 begun to explore the role of this protein in skeletal muscle. We have used co-immunoprecipitation and mass spectrometry to identify proteins that interact with PABPN1 in mouse skeletal muscles. Among the interacting proteins we identified Matrin 3 (MATR3) as a novel protein interactor of PABPN1. The MATR3 gene is mutated in a form of distal myopathy and amyotrophic lateral sclerosis (ALS). We demonstrate, that like PABPN1, MATR3 is critical for myogenesis. Furthermore, MATR3 controls critical aspects of RNA processing including alternative polyadenylation and intron retention. We provide evidence that MATR3 also binds and regulates the levels of long non-coding RNA (lncRNA) Neat1 and together with PABPN1 is required for normal paraspeckle function. We demonstrate that PABPN1 and MATR3 are required for paraspeckles, as well as for adenosine to inosine (A to I) RNA editing of Ctn RNA in muscle cells. We provide a functional link between PABPN1 and MATR3 through regulation of a common lncRNA target with downstream impact on paraspeckle morphology and function. We extend our analysis to a mouse model of OPMD and demonstrate altered paraspeckle morphology in the presence of endogenous levels of alanine-expanded PABPN1. In this study, we report protein-binding partners of PABPN1, which could provide insight into novel functions of PABPN1 in skeletal muscle and identify proteins that could be sequestered with alanine-expanded PABPN1 in the nuclear aggregates found in OPMD.


Assuntos
Músculo Esquelético/metabolismo , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteína I de Ligação a Poli(A)/metabolismo , Processamento Pós-Transcricional do RNA , Proteínas de Ligação a RNA/metabolismo , Animais , Células Cultivadas , Humanos , Camundongos Endogâmicos C57BL , Desenvolvimento Muscular , Proteína I de Ligação a Poli(A)/fisiologia
6.
Traffic ; 17(7): 803-14, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27061800

RESUMO

A well-written application for funding in support of basic biological or biomedical research or individual training fellowship requires that the author perform several functions well. They must (i) identify an important topic, (ii) provide a brief but persuasive introduction to highlight its significance, (iii) identify one or two key questions that if answered would impact the field, (iv) present a series of logical experiments and convince the reader that the approaches are feasible, doable within a certain period of time and have the potential to answer the questions posed, and (v) include citations that demonstrate both scholarship and an appropriate command of the relevant literature and techniques involved in the proposed research study. In addition, preparation of any compelling application requires formal scientific writing and editing skills that are invaluable in any career. These are also all key components in a doctoral dissertation and encompass many of the skills that we expect graduate students to master. Almost 20 years ago, we began a grant writing course as a mechanism to train students in these specific skills. Here, we describe the use of this course in training of our graduate students as well as our experiences and lessons learned.


Assuntos
Pesquisa Biomédica/educação , Currículo , Educação de Pós-Graduação/métodos , Organização do Financiamento , Modelos Educacionais , Redação , Pesquisa Biomédica/economia , Humanos , Projetos de Pesquisa , Estudantes
7.
Am J Physiol Cell Physiol ; 313(4): C392-C404, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28701357

RESUMO

Cell-cell adhesion molecules play key roles in maintaining quiescence or promoting activation of various stem cells in their niche. Muscle stem cells called satellite cells (SC) are critical for skeletal muscle regeneration after injury, but little is known about the role of adhesion molecules in regulating the behavior of these stem cells. Vascular cell adhesion molecule-1 (VCAM-1) is a cell-cell adhesion protein expressed on quiescent and activated SC whose function is unknown in this context. We deleted Vcam1 from SC using an inducible Cre recombinase in young mice. In the injured niche, Vcam1-/- SC underwent premature lineage progression to a more differentiated state as well as apoptosis leading to a transient delay in myofiber growth during regeneration. Apoptosis was also increased in Vcam1-/- SC in vitro concomitant with decreased levels of phosphorylated Akt, a prosurvival signal activated by VCAM-1 signaling in other cell types. During muscle regeneration, we observed an influx of immune cells expressing α4 integrin, a component of the major, high-affinity VCAM-1 ligand, α4ß1 integrin. Furthermore, α4 integrin mRNA and protein were induced in SC 2 days after injury. These results suggest that SC interact with other SC as well as immune cells through α4ß1 integrin in the injured niche to promote expansion of SC. In the uninjured niche, multiple cell types also expressed α4 integrin. However, only basal fusion of Vcam1-/- SC with myofibers was decreased, contributing to decreased myofiber growth. These studies define differential roles for VCAM-1 in SC depending on the state of their niche.


Assuntos
Músculo Esquelético/lesões , Músculo Esquelético/fisiopatologia , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Nicho de Células-Tronco , Molécula 1 de Adesão de Célula Vascular/metabolismo , Animais , Sobrevivência Celular , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Molécula 1 de Adesão de Célula Vascular/genética
8.
Stem Cells ; 34(11): 2784-2797, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27434733

RESUMO

Satellite cells are stem cells with an essential role in skeletal muscle repair. Precise regulation of gene expression is critical for proper satellite cell quiescence, proliferation, differentiation and self-renewal. Nuclear proteins required for gene expression are dependent on the nucleocytoplasmic transport machinery to access to nucleus, however little is known about regulation of nuclear transport in satellite cells. The best characterized nuclear import pathway is classical nuclear import which depends on a classical nuclear localization signal (cNLS) in a cargo protein and the heterodimeric import receptors, karyopherin alpha (KPNA) and beta (KPNB). Multiple KPNA1 paralogs exist and can differ in importing specific cNLS proteins required for cell differentiation and function. We show that transcripts for six Kpna paralogs underwent distinct changes in mouse satellite cells during muscle regeneration accompanied by changes in cNLS proteins in nuclei. Depletion of KPNA1, the most dramatically altered KPNA, caused satellite cells in uninjured muscle to prematurely activate, proliferate and undergo apoptosis leading to satellite cell exhaustion with age. Increased proliferation of satellite cells led to enhanced muscle regeneration at early stages of regeneration. In addition, we observed impaired nuclear localization of two key KPNA1 cargo proteins: p27, a cyclin-dependent kinase inhibitor associated with cell cycle control and lymphoid enhancer factor 1, a critical cotranscription factor for ß-catenin. These results indicate that regulated nuclear import of proteins by KPNA1 is critical for satellite cell proliferation and survival and establish classical nuclear import as a novel regulatory mechanism for controlling satellite cell fate. Stem Cells 2016;34:2784-2797.


Assuntos
Núcleo Celular/metabolismo , Músculo Esquelético/metabolismo , Sinais de Localização Nuclear/genética , Regeneração/genética , Células Satélites de Músculo Esquelético/metabolismo , alfa Carioferinas/genética , Transporte Ativo do Núcleo Celular/genética , Animais , Compostos de Bário/toxicidade , Proliferação de Células , Sobrevivência Celular , Cloretos/toxicidade , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Citoplasma/metabolismo , Feminino , Regulação da Expressão Gênica , Fator 1 de Ligação ao Facilitador Linfoide/genética , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/lesões , Sinais de Localização Nuclear/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Satélites de Músculo Esquelético/citologia , Transdução de Sinais , alfa Carioferinas/deficiência , beta Catenina/genética , beta Catenina/metabolismo
9.
Stem Cells ; 33(12): 3581-95, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26178867

RESUMO

The pharyngeal muscles of the nasal, oral, and laryngeal pharynxes are required for swallowing. Pharyngeal muscles are preferentially affected in some muscular dystrophies yet spared in others. Muscle stem cells, called satellite cells, may be critical factors in the development of pharyngeal muscle disorders; however, very little is known about pharyngeal satellite cells (PSC) and their role in pharyngeal muscles. We show that PSC are distinct from the commonly studied hindlimb satellite cells both transcriptionally and biologically. Under basal conditions PSC proliferate, progress through myogenesis, and fuse with pharyngeal myofibers. Furthermore, PSC exhibit biologic differences dependent on anatomic location in the pharynx. Importantly, PSC are required to maintain myofiber size and myonuclear number in pharyngeal myofibers. Together, these results demonstrate that PSC are critical for pharyngeal muscle maintenance and suggest that satellite cell impairment could contribute to pharyngeal muscle pathology associated with various muscular dystrophies and aging.


Assuntos
Desenvolvimento Muscular , Músculos Faríngeos/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Animais , Camundongos , Camundongos Mutantes , Músculos Faríngeos/citologia , Células Satélites de Músculo Esquelético/citologia
10.
Am J Physiol Cell Physiol ; 308(11): C919-31, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25810257

RESUMO

Myoblast fusion is critical for proper muscle growth and regeneration. During myoblast fusion, the localization of some molecules is spatially restricted; however, the exact reason for such localization is unknown. Creatine kinase B (CKB), which replenishes local ATP pools, localizes near the ends of cultured primary mouse myotubes. To gain insights into the function of CKB, we performed a yeast two-hybrid screen to identify CKB-interacting proteins. We identified molecules with a broad diversity of roles, including actin polymerization, intracellular protein trafficking, and alternative splicing, as well as sarcomeric components. In-depth studies of α-skeletal actin and α-cardiac actin, two predominant muscle actin isoforms, demonstrated their biochemical interaction and partial colocalization with CKB near the ends of myotubes in vitro. In contrast to other cell types, specific knockdown of CKB did not grossly affect actin polymerization in myotubes, suggesting other muscle-specific roles for CKB. Interestingly, knockdown of CKB resulted in significantly increased myoblast fusion and myotube size in vitro, whereas knockdown of creatine kinase M had no effect on these myogenic parameters. Our results suggest that localized CKB plays a key role in myotube formation by limiting myoblast fusion during myogenesis.


Assuntos
Creatina Quinase Forma BB/genética , Desenvolvimento Muscular/genética , Fibras Musculares Esqueléticas/enzimologia , Mioblastos/enzimologia , Actinas/genética , Actinas/metabolismo , Processamento Alternativo , Animais , Fusão Celular , Creatina Quinase Forma BB/antagonistas & inibidores , Creatina Quinase Forma BB/metabolismo , Creatina Quinase Forma MM/genética , Creatina Quinase Forma MM/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/citologia , Mioblastos/citologia , Polimerização , Cultura Primária de Células , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido
11.
Development ; 139(4): 641-56, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22274696

RESUMO

The fusion of myoblasts into multinucleate syncytia plays a fundamental role in muscle function, as it supports the formation of extended sarcomeric arrays, or myofibrils, within a large volume of cytoplasm. Principles learned from the study of myoblast fusion not only enhance our understanding of myogenesis, but also contribute to our perspectives on membrane fusion and cell-cell fusion in a wide array of model organisms and experimental systems. Recent studies have advanced our views of the cell biological processes and crucial proteins that drive myoblast fusion. Here, we provide an overview of myoblast fusion in three model systems that have contributed much to our understanding of these events: the Drosophila embryo; developing and regenerating mouse muscle; and cultured rodent muscle cells.


Assuntos
Fusão Celular , Drosophila/embriologia , Camundongos/embriologia , Mioblastos/fisiologia , Animais , Adesão Celular/fisiologia , Comunicação Celular/fisiologia , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Extensões da Superfície Celular/metabolismo , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Drosophila/anatomia & histologia , Células Gigantes/citologia , Células Gigantes/fisiologia , Fusão de Membrana/fisiologia , Camundongos/anatomia & histologia , Morfogênese/fisiologia , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Mioblastos/citologia , Regeneração/fisiologia , Transdução de Sinais/fisiologia
12.
Dev Biol ; 382(1): 160-71, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23872330

RESUMO

Actin dynamics are necessary at multiple steps in the formation of multinucleated muscle cells. BAR domain proteins can regulate actin dynamics in several cell types, but have been little studied in skeletal muscle. Here, we identify novel functions for the N-BAR domain protein, Bridging integrator 3 (Bin3), during myogenesis in mice. Bin3 plays an important role in regulating myofiber size in vitro and in vivo. During early myogenesis, Bin3 promotes migration of differentiated muscle cells, where it colocalizes with F-actin in lamellipodia. In addition, Bin3 forms a complex with Rac1 and Cdc42, Rho GTPases involved in actin polymerization, which are known to be essential for myotube formation. Importantly, a Bin3-dependent pathway is a major regulator of Rac1 and Cdc42 activity in differentiated muscle cells. Overall, these data classify N-BAR domain proteins as novel regulators of actin-dependent processes in myogenesis, and further implicate BAR domain proteins in muscle growth and repair.


Assuntos
Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/metabolismo , Desenvolvimento Muscular , Neuropeptídeos/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Actinas/metabolismo , Animais , Movimento Celular , Endocitose , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/fisiologia , Estrutura Terciária de Proteína , Pseudópodes/metabolismo , Regeneração
13.
Stem Cells ; 31(2): 384-96, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23169615

RESUMO

Macrophages (MPs) exert either beneficial or deleterious effects on tissue repair, depending on their activation/polarization state. They are crucial for adult skeletal muscle repair, notably by acting on myogenic precursor cells. However, these interactions have not been fully characterized. Here, we explored both in vitro and in vivo, in human, the interactions of differentially activated MPs with myogenic precursor cells (MPCs) during adult myogenesis and skeletal muscle regeneration. We showed in vitro that through the differential secretion of cytokines and growth factors, proinflammatory MPs inhibited MPC fusion while anti-inflammatory MPs strongly promoted MPC differentiation by increasing their commitment into differentiated myocytes and the formation of mature myotubes. Furthermore, the in vivo time course of expression of myogenic and MP markers was studied in regenerating human healthy muscle after damage. We observed that regenerating areas containing proliferating MPCs were preferentially associated with MPs expressing proinflammatory markers. In the same muscle, regenerating areas containing differentiating myogenin-positive MPCs were preferentially coupled to MPs harboring anti-inflammatory markers. These data demonstrate for the first time in human that MPs sequentially orchestrate adult myogenesis during regeneration of damaged skeletal muscle. These results support the emerging concept that inflammation, through MP activation, controls stem cell fate and coordinates tissue repair.


Assuntos
Células-Tronco Adultas/citologia , Macrófagos/citologia , Fibras Musculares Esqueléticas/citologia , Músculo Esquelético/citologia , Regeneração/fisiologia , Adulto , Células-Tronco Adultas/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular , Células Cultivadas , Citocinas/biossíntese , Citocinas/metabolismo , Expressão Gênica , Humanos , Inflamação , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Ativação de Macrófagos , Macrófagos/classificação , Macrófagos/metabolismo , Desenvolvimento Muscular/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Miogenina/genética , Miogenina/metabolismo
14.
Dev Biol ; 357(1): 248-58, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21741962

RESUMO

Proper muscle function is dependent on spatial and temporal control of gene expression in myofibers. Myofibers are multinucleated cells that are formed, repaired and maintained by the process of myogenesis in which progenitor myoblasts proliferate, differentiate and fuse. Gene expression is dependent upon proteins that require facilitated nuclear import, however little is known about the regulation of nucleocytoplasmic transport during the formation of myofibers. We analyzed the role of karyopherin alpha (KPNA), a key classical nuclear import receptor, during myogenesis. We established that five karyopherin alpha paralogs are expressed by primary mouse myoblasts in vitro and that their steady-state levels increase in multinucleated myotubes, suggesting a global increase in demand for classical nuclear import during myogenesis. We used siRNA-mediated knockdown to identify paralog-specific roles for KPNA1 and KPNA2 during myogenesis. KPNA1 knockdown increased myoblast proliferation, whereas KPNA2 knockdown decreased proliferation. In contrast, no proliferation defect was observed with KPNA4 knockdown. Only knockdown of KPNA2 decreased myotube growth. These results identify distinct pathways involved in myoblast proliferation and myotube growth that rely on specific nuclear import receptors suggesting that regulation of classical nuclear import pathways likely plays a critical role in controlling gene expression in skeletal muscle.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Células Musculares/metabolismo , Desenvolvimento Muscular , Proteínas Nucleares/metabolismo , alfa Carioferinas/metabolismo , Animais , Diferenciação Celular , Movimento Celular , Proliferação de Células , Camundongos , Camundongos Endogâmicos BALB C , Células Musculares/citologia , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Proteínas Nucleares/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , alfa Carioferinas/genética
15.
Hum Mol Genet ; 19(6): 1058-65, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20035013

RESUMO

The nuclear poly(A)-binding protein 1 (PABPN1) is a ubiquitously expressed protein that plays a critical role in polyadenylation. Short expansions of the polyalanine tract in the N-terminus of PABPN1 lead to oculopharyngeal muscular dystrophy (OPMD), which is an adult onset disease characterized by eyelid drooping, difficulty in swallowing and weakness in the proximal limb muscles. Although significant data from in vitro biochemical assays define the function of PABPN1 in control of poly(A) tail length, little is known about the role of PABPN1 in mammalian cells. To assess the function of PABPN1 in mammalian cells and specifically in cells affected in OPMD, we examined the effects of PABPN1 depletion using siRNA in primary mouse myoblasts from extraocular, pharyngeal and limb muscles. PABPN1 knockdown significantly decreased cell proliferation and myoblast differentiation during myogenesis in vitro. At the molecular level, PABPN1 depletion in myoblasts led to a shortening of mRNA poly(A) tails, demonstrating the cellular function of PABPN1 in polyadenylation control in a mammalian cell. In addition, PABPN1 depletion caused nuclear accumulation of poly(A) RNA, revealing that PABPN1 is required for proper poly(A) RNA export from the nucleus. Together, these experiments demonstrate that PABPN1 plays an essential role in myoblast proliferation and differentiation, suggesting that it is required for muscle regeneration and maintenance in vivo.


Assuntos
Núcleo Celular/metabolismo , Desenvolvimento Muscular , Proteína II de Ligação a Poli(A)/metabolismo , Proteína I de Ligação a Poli(A)/metabolismo , RNA Mensageiro/biossíntese , Animais , Diferenciação Celular , Proliferação de Células , Camundongos , Camundongos Endogâmicos BALB C , Mioblastos/citologia , Mioblastos/metabolismo , Poli A/metabolismo , Poliadenilação , Transporte de RNA
16.
J Cell Sci ; 123(Pt 18): 3052-60, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20736301

RESUMO

Adult regenerative myogenesis is vital for restoring normal tissue structure after muscle injury. Muscle regeneration is dependent on progenitor satellite cells, which proliferate in response to injury, and their progeny differentiate and undergo cell-cell fusion to form regenerating myofibers. Myogenic progenitor cells must be precisely regulated and positioned for proper cell fusion to occur. Chemokines are secreted proteins that share both leukocyte chemoattractant and cytokine-like behavior and affect the physiology of a number of cell types. We investigated the steady-state mRNA levels of 84 chemokines, chemokine receptors and signaling molecules, to obtain a comprehensive view of chemokine expression by muscle cells during myogenesis in vitro. A large number of chemokines and chemokine receptors were expressed by primary mouse muscle cells, especially during times of extensive cell-cell fusion. Furthermore, muscle cells exhibited different migratory behavior throughout myogenesis in vitro. One receptor-ligand pair, CXCR4-SDF-1alpha (CXCL12), regulated migration of both proliferating and terminally differentiated muscle cells, and was necessary for proper fusion of muscle cells. Given the large number of chemokines and chemokine receptors directly expressed by muscle cells, these proteins might have a greater role in myogenesis than previously appreciated.


Assuntos
Movimento Celular , Quimiocinas/genética , Regulação da Expressão Gênica , Desenvolvimento Muscular , Mioblastos/citologia , Animais , Células Cultivadas , Quimiocinas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mioblastos/metabolismo , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo
17.
FASEB J ; 25(4): 1156-65, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21156809

RESUMO

Sca-1 (stem cell antigen-1) is a member of the Ly-6 family of proteins and regulates cell proliferation, differentiation, and self-renewal in multiple tissues. In skeletal muscle, Sca-1 inhibits both proliferation and differentiation of myogenic cells. Sca-1 expression is dynamically regulated during muscle regeneration, and mice lacking Sca-1 display increased fibrosis following muscle injury. Here, we show that Sca-1 expression is negatively regulated by TGF-ß1 and that this inhibition is dependent on Smad3. We demonstrate that levels of TGF-ß1 in skeletal muscle rapidly increase on injury and that the majority of this TGFß1 is produced by infiltrating macrophages. Sca-1 is expressed in multiple cell types, and we demonstrate that TGF-ß1 represses Sca-1 expression in T cells and other immune cell populations derived from the spleen, indicating that regulation by TGF-ß1 is a general feature of Sca-1 expression in multiple cell types. Elucidation of the mechanisms by which Sca-1 expression is regulated may aid in the understanding of muscle homeostasis, potentially identifying novel therapeutic targets for muscle diseases.


Assuntos
Antígenos Ly/biossíntese , Proteínas de Membrana/biossíntese , Músculo Esquelético/fisiologia , Mioblastos Esqueléticos/fisiologia , Regeneração/fisiologia , Fator de Crescimento Transformador beta1/fisiologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Proteína Smad3/fisiologia
18.
Proc Natl Acad Sci U S A ; 106(12): 4719-24, 2009 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-19264965

RESUMO

Most cells in adult tissues are nondividing. In skeletal muscle, differentiated myofibers have exited the cell cycle permanently, whereas satellite stem cells withdraw transiently, returning to active proliferation to repair damaged myofibers. We have examined the epigenetic mechanisms operating in conditional quiescence by analyzing the function of a predicted chromatin regulator mixed lineage leukemia 5 (MLL5) in a culture model of reversible arrest. MLL5 is induced in quiescent myoblasts and regulates both the cell cycle and differentiation via a hierarchy of chromatin and transcriptional regulators. Knocking down MLL5 delays entry of quiescent myoblasts into S phase, but hastens S-phase completion. Cyclin A2 (CycA) mRNA is no longer restricted to S phase, but is induced throughout G(0)/G(1), with activation of the cell cycle regulated element (CCRE) in the CycA promoter. Overexpressed MLL5 physically associates with the CCRE and impairs its activity. MLL5 also regulates CycA indirectly: Cux, an activator of CycA promoter and S phase is induced in RNAi cells, and Brm/Brg1, CCRE-binding repressors that promote differentiation are repressed. In knockdown cells, H3K4 methylation at the CCRE is reduced, reflecting quantitative global changes in methylation. MLL5 appears to lack intrinsic histone methyl transferase activity, but regulates expression of histone-modifying enzymes LSD1 and SET7/9, suggesting an indirect mechanism. Finally, expression of muscle regulators Pax7, Myf5, and myogenin is impaired in MLL5 knockdown cells, which are profoundly differentiation defective. Collectively, our results suggest that MLL5 plays an integral role in novel chromatin regulatory mechanisms that suppress inappropriate expression of S-phase-promoting genes and maintain expression of determination genes in quiescent cells.


Assuntos
Diferenciação Celular , Proteínas Cromossômicas não Histona/química , Ciclina A/metabolismo , Proteínas de Drosophila/química , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Mioblastos/citologia , Animais , Proteínas Cromossômicas não Histona/metabolismo , Ciclina A/genética , Ciclina A2 , Fase G1 , Técnicas de Silenciamento de Genes , Histona-Lisina N-Metiltransferase/deficiência , Metilação , Camundongos , Mioblastos/enzimologia , Ligação Proteica , Interferência de RNA , Proteínas Repressoras/metabolismo , Elementos de Resposta/genética , Fase S , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/metabolismo , Transcrição Gênica
19.
Am J Physiol Cell Physiol ; 300(2): C287-94, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21123737

RESUMO

Efficient muscle regeneration requires the clearance of dead and dying tissue via phagocytosis before remodeling. We have previously shown that mice lacking stem cell antigen-1 (Sca-1) display a defect in skeletal muscle regeneration characterized by increased fibrosis and decreased turnover of the extracellular matrix. In the present study we demonstrate that Sca-1(-/-) mice have a defect in their capacity to recruit soluble IgM, and subsequently C3 complement, to damaged muscle. We hypothesize that this defect in recruitment delays or decreases phagocytosis by macrophages, contributing to the previously observed fibrotic phenotype of these mice. As the primary source of soluble IgM is peritoneal B-1a cells, which are a subset of self-renewing B cells, we analyzed this cell population and observed a significant reduction in B-1a cells in Sca-1(-/-) animals. Interestingly, these mice are protected from ischemia-reperfusion injury, an acute inflammatory reaction also mediated by IgM and C3 complement that has been linked to a deficit in B-1a cells in previous studies. Collectively, these data reveal a novel role for Sca-1 in innate immunity during muscle regeneration and indicate that further elucidation of immuno-myogenic processes will help to better understand and promote muscle regeneration.


Assuntos
Antígenos Ly/imunologia , Imunidade Inata , Proteínas de Membrana/imunologia , Músculo Esquelético/imunologia , Regeneração/imunologia , Animais , Antígenos Ly/genética , Subpopulações de Linfócitos B/imunologia , Complemento C3/imunologia , Feminino , Imunoglobulina M/imunologia , Macrófagos/imunologia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Músculo Esquelético/fisiologia , Fagocitose/imunologia , Regeneração/genética , Regeneração/fisiologia , Traumatismo por Reperfusão/imunologia
20.
J Cell Sci ; 122(Pt 19): 3481-91, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19723804

RESUMO

Quiescent muscle progenitors called satellite cells persist in adult skeletal muscle and, upon injury to muscle, re-enter the cell cycle and either undergo self-renewal or differentiate to regenerate lost myofibers. Using synchronized cultures of C2C12 myoblasts to model these divergent programs, we show that p8 (also known as Nupr1), a G1-induced gene, negatively regulates the cell cycle and promotes myogenic differentiation. p8 is a small chromatin protein related to the high mobility group (HMG) family of architectural factors and binds to histone acetyltransferase p300 (p300, also known as CBP). We confirm this interaction and show that p300-dependent events (Myc expression, global histone acetylation and post-translational acetylation of the myogenic regulator MyoD) are all affected in p8-knockdown myoblasts, correlating with repression of MyoD target-gene expression and severely defective differentiation. We report two new partners for p8 that support a role in muscle-specific gene regulation: p68 (Ddx5), an RNA helicase reported to bind both p300 and MyoD, and MyoD itself. We show that, similar to MyoD and p300, p8 and p68 are located at the myogenin promoter, and that knockdown of p8 compromises chromatin association of all four proteins. Thus, p8 represents a new node in a chromatin regulatory network that coordinates myogenic differentiation with cell-cycle exit.


Assuntos
Cromatina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Substâncias de Crescimento/metabolismo , Mioblastos/citologia , Miogenina/genética , Proteínas de Neoplasias/metabolismo , Regiões Promotoras Genéticas , Animais , Ciclo Celular , Diferenciação Celular , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteína p300 Associada a E1A/genética , Proteína p300 Associada a E1A/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Substâncias de Crescimento/genética , Humanos , Camundongos , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Proteína MyoD/genética , Proteína MyoD/metabolismo , Mioblastos/metabolismo , Miogenina/metabolismo , Proteínas de Neoplasias/genética , Ligação Proteica , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA