Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nucleic Acids Res ; 51(12): 5901-5910, 2023 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-37224533

RESUMO

Although targeting TfR1 to deliver oligonucleotides to skeletal muscle has been demonstrated in rodents, effectiveness and pharmacokinetic/pharmacodynamic (PKPD) properties remained unknown in higher species. We developed antibody-oligonucleotide conjugates (AOCs) towards mice or monkeys utilizing anti-TfR1 monoclonal antibodies (αTfR1) conjugated to various classes of oligonucleotides (siRNA, ASOs and PMOs). αTfR1 AOCs delivered oligonucleotides to muscle tissue in both species. In mice, αTfR1 AOCs achieved a > 15-fold higher concentration to muscle tissue than unconjugated siRNA. A single dose of an αTfR1 conjugated to an siRNA against Ssb mRNA produced > 75% Ssb mRNA reduction in mice and monkeys, and mRNA silencing was greatest in skeletal and cardiac (striated) muscle with minimal to no activity in other major organs. In mice the EC50 for Ssb mRNA reduction in skeletal muscle was >75-fold less than in systemic tissues. Oligonucleotides conjugated to control antibodies or cholesterol produced no mRNA reduction or were 10-fold less potent, respectively. Tissue PKPD of AOCs demonstrated mRNA silencing activity primarily driven by receptor-mediated delivery in striated muscle for siRNA oligonucleotides. In mice, we show that AOC-mediated delivery is operable across various oligonucleotide modalities. AOC PKPD properties translated to higher species, providing promise for a new class of oligonucleotide therapeutics.


Assuntos
Oligonucleotídeos Antissenso , Oligonucleotídeos , Camundongos , Animais , Anticorpos/uso terapêutico , RNA Interferente Pequeno/genética , RNA Mensageiro/genética , Músculo Esquelético
2.
EMBO Rep ; 16(8): 1037-50, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26136374

RESUMO

Although the two catalytic subunits of the SWI/SNF chromatin-remodeling complex--Brahma (Brm) and Brg1--are almost invariably co-expressed, their mutually exclusive incorporation into distinct SWI/SNF complexes predicts that Brg1- and Brm-based SWI/SNF complexes execute specific functions. Here, we show that Brg1 and Brm have distinct functions at discrete stages of muscle differentiation. While Brg1 is required for the activation of muscle gene transcription at early stages of differentiation, Brm is required for Ccnd1 repression and cell cycle arrest prior to the activation of muscle genes. Ccnd1 knockdown rescues the ability to exit the cell cycle in Brm-deficient myoblasts, but does not recover terminal differentiation, revealing a previously unrecognized role of Brm in the activation of late muscle gene expression independent from the control of cell cycle. Consistently, Brm null mice displayed impaired muscle regeneration after injury, with aberrant proliferation of satellite cells and delayed formation of new myofibers. These data reveal stage-specific roles of Brm during skeletal myogenesis, via formation of repressive and activatory SWI/SNF complexes.


Assuntos
Pontos de Checagem do Ciclo Celular/genética , DNA Helicases/metabolismo , Expressão Gênica , Desenvolvimento Muscular/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Ciclina D1/deficiência , Ciclina D1/genética , DNA Helicases/genética , Técnicas de Silenciamento de Genes , Camundongos , Células Musculares , Proteínas Nucleares/genética , Fatores de Transcrição/genética
3.
EMBO J ; 31(2): 301-16, 2012 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-22068056

RESUMO

Tissue-specific transcriptional activators initiate differentiation towards specialized cell types by inducing chromatin modifications permissive for transcription at target loci, through the recruitment of SWItch/Sucrose NonFermentable (SWI/SNF) chromatin-remodelling complex. However, the molecular mechanism that regulates SWI/SNF nuclear distribution in response to differentiation signals is unknown. We show that the muscle determination factor MyoD and the SWI/SNF subunit BAF60c interact on the regulatory elements of MyoD-target genes in myoblasts, prior to activation of transcription. BAF60c facilitates MyoD binding to target genes and marks the chromatin for signal-dependent recruitment of the SWI/SNF core to muscle genes. BAF60c phosphorylation on a conserved threonine by differentiation-activated p38α kinase is the signal that promotes incorporation of MyoD-BAF60c into a Brg1-based SWI/SNF complex, which remodels the chromatin and activates transcription of MyoD-target genes. Our data support an unprecedented two-step model by which pre-assembled BAF60c-MyoD complex directs recruitment of SWI/SNF to muscle loci in response to differentiation cues.


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Sistema de Sinalização das MAP Quinases , Desenvolvimento Muscular/fisiologia , Proteínas Musculares/fisiologia , Proteína MyoD/fisiologia , Fatores de Transcrição/fisiologia , Animais , Linhagem Celular , Cromatina/genética , Proteínas Cromossômicas não Histona/antagonistas & inibidores , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/genética , DNA Helicases/fisiologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica/genética , Células HeLa/metabolismo , Humanos , Camundongos , Complexos Multiproteicos , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/química , Proteínas Musculares/genética , Mioblastos/metabolismo , Proteínas Nucleares/fisiologia , Fosforilação , Fosfotreonina/análise , Processamento de Proteína Pós-Traducional , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/química , Fatores de Transcrição/genética , Técnicas do Sistema de Duplo-Híbrido , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
4.
EMBO J ; 35(15): 1600-2, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27302090

Assuntos
Coração , Músculos , Humanos
5.
Circ Res ; 112(2): 257-66, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23168335

RESUMO

RATIONALE: Nkx2.5 is a transcription factor that regulates cardiomyogenesis in vivo and in embryonic stem cells. It is also a common target in congenital heart disease. Although Nkx2.5 has been implicated in the regulation of many cellular processes that ultimately contribute to cardiomyogenesis and morphogenesis of the mature heart, relatively little is known about how it is regulated at a functional level. OBJECTIVE: We have undertaken a proteomic screen to identify novel binding partners of Nkx2.5 during cardiomyogenic differentiation in an effort to better understand the regulation of its transcriptional activity. METHODS AND RESULTS: Purification of Nkx2.5 from differentiating cells identified the myosin phosphatase subunits protein phosphatase 1ß and myosin phosphatase targeting subunit 1 (Mypt1) as novel binding partners. The interaction with protein phosphatase 1 ß/Mypt1 resulted in exclusion of Nkx2.5 from the nucleus and, consequently, inhibition of its transcriptional activity. Exclusion of Nkx2.5 was inhibited by treatment with leptomycin B and was dependent on an Mypt1 nuclear export signal. Furthermore, in transient transfection experiments, Nkx2.5 colocalized outside the nucleus with phosphorylated Mypt1 in a manner dependent on Wnt signaling and Rho-associated protein kinase. Treatment of differentiating mouse embryonic stem cells with Wnt3a resulted in enhanced phosphorylation of endogenous Mypt1, increased nuclear exclusion of endogenous Nkx2.5, and a failure to undergo terminal cardiomyogenesis. Finally, knockdown of Mypt1 resulted in rescue of Wnt3a-mediated inhibition of cardiomyogenesis, indicating that Mypt1 is required for this process. CONCLUSIONS: We have identified a novel interaction between Nkx2.5 and myosin phosphatase. Promoting this interaction represents a novel mechanism whereby Wnt3a regulates Nkx2.5 and inhibits cardiomyogenesis.


Assuntos
Inibidores do Crescimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Miócitos Cardíacos/fisiologia , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt/fisiologia , Proteína Wnt3A/fisiologia , Quinases Associadas a rho/fisiologia , Animais , Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/fisiologia , Células HEK293 , Proteína Homeobox Nkx-2.5 , Humanos , Camundongos , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Proteína Fosfatase 1/metabolismo , Frações Subcelulares/enzimologia , Frações Subcelulares/metabolismo
6.
Nat Commun ; 9(1): 3670, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30202063

RESUMO

Fibro-adipogenic progenitors (FAPs) are currently defined by their anatomical position, expression of non-specific membrane-associated proteins, and ability to adopt multiple lineages in vitro. Gene expression analysis at single-cell level reveals that FAPs undergo dynamic transitions through a spectrum of cell states that can be identified by differential expression levels of Tie2 and Vcam1. Different patterns of Vcam1-negative Tie2high or Tie2low and Tie2low/Vcam1-expressing FAPs are detected during neonatal myogenesis, response to acute injury and Duchenne Muscular Dystrophy (DMD). RNA sequencing analysis identified cell state-specific transcriptional profiles that predict functional interactions with satellite and inflammatory cells. In particular, Vcam1-expressing FAPs, which exhibit a pro-fibrotic expression profile, are transiently activated by acute injury in concomitance with the inflammatory response. Aberrant persistence of Vcam1-expressing FAPs is detected in DMD muscles or upon macrophage depletion, and is associated with muscle fibrosis, thereby revealing how disruption of inflammation-regulated FAPs dynamics leads to a pathogenic outcome.


Assuntos
Adipogenia/fisiologia , Desenvolvimento Muscular/fisiologia , Distrofia Muscular de Duchenne/metabolismo , Células-Tronco/metabolismo , Animais , Diferenciação Celular , Citometria de Fluxo , Perfilação da Expressão Gênica , Inflamação , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Endogâmicos mdx , Músculo Esquelético/fisiologia , Receptor TIE-2/metabolismo , Regeneração , Análise de Sequência de RNA , Molécula 1 de Adesão de Célula Vascular/metabolismo
7.
Methods Mol Biol ; 1556: 191-219, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28247351

RESUMO

Single cell gene expression profiling is a fundamental tool for studying the heterogeneity of a cell population by addressing the phenotypic and functional characteristics of each cell. Technological advances that have coupled microfluidic technologies with high-throughput quantitative RT-PCR analyses have enabled detailed analyses of single cells in various biological contexts. In this chapter, we describe the procedure for isolating the skeletal muscle interstitial cells termed Fibro-Adipogenic Progenitors (FAPs ) and their gene expression profiling at the single cell level. Moreover, we accompany our bench protocol with bioinformatics analysis designed to process raw data as well as to visualize single cell gene expression data. Single cell gene expression profiling is therefore a useful tool in the investigation of FAPs heterogeneity and their contribution to muscle homeostasis.


Assuntos
Perfilação da Expressão Gênica , Desenvolvimento Muscular/genética , Músculo Esquelético/citologia , Análise de Célula Única , Animais , Biomarcadores , Técnicas de Cultura de Células , Separação Celular/métodos , Análise por Conglomerados , Biologia Computacional/métodos , Citometria de Fluxo/métodos , Expressão Gênica , Perfilação da Expressão Gênica/métodos , Imunofenotipagem/métodos , Camundongos , Controle de Qualidade , Reação em Cadeia da Polimerase em Tempo Real , Análise de Célula Única/métodos , Software , Estatística como Assunto , Transcriptoma , Navegador
8.
PLoS One ; 12(6): e0179464, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28609469

RESUMO

Post-translational modifications of histones play a key role in the regulation of gene expression during development and differentiation. Numerous studies have shown the dynamics of combinatorial regulation by transcription factors and histone modifications, in the sense that different combinations lead to distinct expression outcomes. Here, we investigated gene regulation by stable enrichment patterns of histone marks H3K4me2 and H3K4me3 in combination with the chromatin binding of the muscle tissue-specific transcription factor MyoD during myogenic differentiation of C2C12 cells. Using k-means clustering, we found that specific combinations of H3K4me2/3 profiles over and towards the gene body impact on gene expression and marks a subset of genes important for muscle development and differentiation. By further analysis, we found that the muscle key regulator MyoD was significantly enriched on this subset of genes and played a repressive role during myogenic differentiation. Among these genes, we identified the pluripotency gene Patz1, which is repressed during myogenic differentiation through direct binding of MyoD to promoter elements. These results point to the importance of integrating histone modifications and MyoD chromatin binding for coordinated gene activation and repression during myogenic differentiation.


Assuntos
Diferenciação Celular/genética , Histonas/genética , Proteína MyoD/genética , Mioblastos/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Análise por Conglomerados , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica/métodos , Ontologia Genética , Células HEK293 , Histonas/classificação , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilação , Camundongos , Desenvolvimento Muscular/genética , Proteína MyoD/metabolismo , Mioblastos/citologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Ligação Proteica , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Elife ; 52016 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-26880551

RESUMO

Change in the identity of the components of the transcription pre-initiation complex is proposed to control cell type-specific gene expression. Replacement of the canonical TFIID-TBP complex with TRF3/TBP2 was reported to be required for activation of muscle-gene expression. The lack of a developmental phenotype in TBP2 null mice prompted further analysis to determine whether TBP2 deficiency can compromise adult myogenesis. We show here that TBP2 null mice have an intact regeneration potential upon injury and that TBP2 is not expressed in established C2C12 muscle cell or in primary mouse MuSCs. While TFIID subunits and TBP are downregulated during myoblast differentiation, reduced amounts of these proteins form a complex that is detectable on promoters of muscle genes and is essential for their expression. This evidence demonstrates that TBP2 does not replace TBP during muscle differentiation, as previously proposed, with limiting amounts of TFIID-TBP being required to promote muscle-specific gene expression.


Assuntos
Regulação da Expressão Gênica , Células Musculares/fisiologia , Proteína MyoD/metabolismo , Proteína de Ligação a TATA-Box/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Camundongos , Camundongos Knockout
10.
Transcription ; 6(2): 21-32, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25996597

RESUMO

TFIIA is an important positive regulator of TFIID, the primary promoter recognition factor of the basal RNA polymerase II transcription machinery. TFIIA antagonises negative TFIID regulators such as negative cofactor 2 (NC2), promotes specific binding of the TBP subunit of TFIID to TATA core promoter sequence elements and stimulates the interaction of TBP-associated factors (TAFs) in the TFIID complex with core promoter elements located downstream of TATA, such as the initiator element (INR). Metazoan TFIIA consists of 3 subunits, TFIIAα (35 kDa), ß (19 kDa) and γ (12 kDa). TFIIAα and ß subunits are encoded by a single gene and result from site-specific cleavage of a 55 kDa TFIIA(α/ß) precursor protein by the protease Taspase1. Metazoan cells have been shown to contain variable amounts of TFIIA (55/12 kDa) and Taspase1-processed TFIIA (35/19/12 kDa) depending on cell type, suggesting distinct gene-specific roles of unprocessed and Taspase1-processed TFIIA. How precisely Taspase1 processing affects TFIIA functions is not understood. Here we report that Taspase1 processing alters TFIIA interactions with TFIID and the conformation of TFIID/TFIIA promoter complexes. We further show that Taspase1 processing induces increased sensitivity of TFIID/TFIIA complexes to the repressor NC2, which is counteracted by the presence of an INR core promoter element. Our results provide first evidence that Taspase1 processing affects TFIIA regulation of TFIID and suggest that Taspase1 processing of TFIIA is required to establish INR-selective core promoter activity in the presence of NC2.


Assuntos
Endopeptidases/metabolismo , Fator de Transcrição TFIIA/biossíntese , Fator de Transcrição TFIID/biossíntese , Proteínas de Ligação a DNA/genética , Endopeptidases/genética , Células HeLa , Humanos , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas , Subunidades Proteicas/genética , RNA Polimerase II/genética , TATA Box/genética , Fatores Associados à Proteína de Ligação a TATA/genética , Fator de Transcrição TFIIA/genética , Fator de Transcrição TFIID/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
11.
Gene ; 303: 165-75, 2003 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-12559578

RESUMO

Mrjp1 gene belongs to the honeybee mrjp gene family encoding the major royal jelly proteins (MRJPs), secreted by nurse bees into the royal jelly. In this study, we have isolated the genomic clone containing the entire mrjp1 gene and determined its sequence. The mrjp1 gene sequence spans over 3038 bp and contains six exons separated by five introns. Seven mismatches between the mrjp1 gene sequence and two previously independently published cDNA sequences were found, but these differences do not lead to any change in the deduced amino acid sequence of MRJP1. With the aid of inverse polymerase chain reaction we obtained sequences flanking the 5' ends of other mrjp genes (mrjp2, mrjp3, mrjp4 and mrjp5). Putative promoters were predicted upstream of all mrjp genes (including mrjp1). The predicted promoters contain the TATA motif (TATATATT), highly conserved both in sequence and position. Ultraspiracle (USP) transcription factor (TF) binding sites in putative promoter regions and clusters of dead ringer TF binding sites upstream of these promoters were predicted computationally. We propose that USP, as a juvenile hormone (JH) binding TF, might possibly act as a mediator of mrjp expression in response to JH. Mrjp1's genomic locus is predicted to encode an antisense transcript, partially overlapping with five mrjp1 exons and entirely overlapping with the putative promoter and predicted transcriptional start point of mrjp1. This finding may shed light on the mechanisms of regulation of mrjps expression. Southern blot analysis of genomic DNA revealed that all so far known members of mrjp gene family (mrjp1, mrjp2, mrjp3, mrjp4 and mrjp5) are present as single-copy genes per haploid honeybee genome. Although MRJPs and the yellow protein of Drosophila melanogaster share a certain degree of similarity in aa sequence and although it has been shown that they share a common evolutionary origin, neither structural similarities in the gene organization, nor significant similarities between intron sequences of mrjp1 gene and fourteen yellow-like genes of D. melanogaster were found.


Assuntos
Abelhas/genética , Proteínas de Insetos/genética , Família Multigênica/genética , Regiões Promotoras Genéticas/genética , Animais , Sequência de Bases , Sítios de Ligação/genética , Southern Blotting , DNA/química , DNA/genética , DNA/metabolismo , Éxons , Feminino , Genes/genética , Íntrons , Larva/genética , Dados de Sequência Molecular , Sequências Reguladoras de Ácido Nucleico/genética , Análise de Sequência de DNA , Fatores de Transcrição/metabolismo
12.
Nat Med ; 20(10): 1182-6, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25194572

RESUMO

The progressive loss of muscle regenerative capacity with age or disease results in part from a decline in the number and function of satellite cells, the direct cellular contributors to muscle repair. However, little is known about the molecular effectors underlying satellite cell impairment and depletion. Elevated levels of inflammatory cytokines, including interleukin-6 (IL-6), are associated with both age-related and muscle-wasting conditions. The levels of STAT3, a downstream effector of IL-6, are also elevated with muscle wasting, and STAT3 has been implicated in the regulation of self-renewal and stem cell fate in several tissues. Here we show that IL-6-activated Stat3 signaling regulates satellite cell behavior, promoting myogenic lineage progression through myogenic differentiation 1 (Myod1) regulation. Conditional ablation of Stat3 in Pax7-expressing satellite cells resulted in their increased expansion during regeneration, but compromised myogenic differentiation prevented the contribution of these cells to regenerating myofibers. In contrast, transient Stat3 inhibition promoted satellite cell expansion and enhanced tissue repair in both aged and dystrophic muscle. The effects of STAT3 inhibition on cell fate and proliferation were conserved in human myoblasts. The results of this study indicate that pharmacological manipulation of STAT3 activity can be used to counteract the functional exhaustion of satellite cells in pathological conditions, thereby maintaining the endogenous regenerative response and ameliorating muscle-wasting diseases.


Assuntos
Fator de Transcrição STAT3/fisiologia , Células Satélites de Músculo Esquelético/fisiologia , Envelhecimento/genética , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Proliferação de Células , Células Cultivadas , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Knockout , Desenvolvimento Muscular/fisiologia , Proteína MyoD/genética , Proteína MyoD/metabolismo , Fator de Transcrição PAX7/metabolismo , Regeneração/fisiologia , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Células Satélites de Músculo Esquelético/citologia , Transdução de Sinais
13.
Cell Rep ; 3(3): 661-70, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23478022

RESUMO

Direct generation of a homogeneous population of skeletal myoblasts from human embryonic stem cells (hESCs) and formation of three-dimensional contractile structures for disease modeling in vitro are current challenges in regenerative medicine. Previous studies reported on the generation of myoblasts from ESC-derived embryoid bodies (EB), but not from undifferentiated ESCs, indicating the requirement for mesodermal transition to promote skeletal myogenesis. Here, we show that selective absence of the SWI/SNF component BAF60C (encoded by SMARCD3) confers on hESCs resistance to MyoD-mediated activation of skeletal myogenesis. Forced expression of BAF60C enables MyoD to directly activate skeletal myogenesis in hESCs by instructing MyoD positioning and allowing chromatin remodeling at target genes. BAF60C/MyoD-expressing hESCs are epigenetically committed myogenic progenitors, which bypass the mesodermal requirement and, when cultured as floating clusters, give rise to contractile three-dimensional myospheres composed of skeletal myotubes. These results identify BAF60C as a key epigenetic determinant of hESC commitment to the myogenic lineage and establish the molecular basis for the generation of hESC-derived myospheres exploitable for "disease in a dish" models of muscular physiology and dysfunction.


Assuntos
Reprogramação Celular , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Fibras Musculares Esqueléticas/citologia , Linhagem Celular , Linhagem da Célula , Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona , Células-Tronco Embrionárias/citologia , Humanos , Mesoderma/citologia , Contração Muscular , Desenvolvimento Muscular/genética , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/fisiologia , Proteína MyoD/genética , Proteína MyoD/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
J Stem Cell Res Ther ; (Suppl 11)2012 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-24634800

RESUMO

The capacity of adult skeletal muscle for regeneration appears to be limited, with progressive impairment in repair efficiency of injured muscles observed in chronic muscular disorders and during aging. While satellite cells, the committed adult muscle stem cells, are the main direct cell source supporting the regenerative potential of adult skeletal muscles, the characterization of the cell types and signals that constitute the functional "niche" of satellite cells is currently the object of intense investigation. Recent studies have identified a functional relationship between satellite cells and various cell types located in key anatomical position, such as the interstitium of skeletal muscles. This heterogeneous population of muscle interstitial cells (MICs) appears to retain an intrinsic multipotency within the mesodermal lineage, and their direct or indirect contribution to myofiber turnover, repair and degeneration has been suggested by many studies that will be reviewed here. Given the existing gap of knowledge on lineage identity and functional properties of MICs, their detailed characterization at the single cell level holds the promise to provide key insight into the composition of this heterogeneous population and the dynamic transition through distinct sub-populations in healthy, diseased and aging muscles. This review provides an overview of the results of various studies describing the phenotype and the function of cells isolated from skeletal muscle interstitium, and discusses the importance of single cell transcription profiling in order to decipher the functional and phenotypical heterogeneity of muscle interstitial cells (MICs).

15.
Curr Opin Mol Ther ; 12(2): 214-22, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20373265

RESUMO

Chromatin remodeling guided by non-coding RNA (ncRNA) contributes mechanistically to the establishment of chromatin structure and to the maintenance of epigenetic memory. Various ncRNAs have been identified as regulators of chromatin structure and gene expression. The widespread occurrence of antisense transcription in eukaryotes emphasizes the prevalence of gene regulation by natural antisense transcripts. Recently, antisense ncRNAs have been implicated in the silencing of tumor suppressor genes through epigenetic remodeling events. Characterization of the antisense RNAs involved in the development or maintenance of oncogenic states may define ncRNAs as early biomarkers for the emergence of cancer, and could have a significant impact on the development of tools for disease diagnosis and treatment. In this review, current knowledge on the mechanisms of ncRNA-mediated transcriptional gene silencing in humans is discussed, and parallels between the establishment of a silent chromatin state mediated by siRNAs and long antisense ncRNAs are highlighted.


Assuntos
Epigênese Genética , Inativação Gênica , RNA não Traduzido , Transcrição Gênica , Animais , Montagem e Desmontagem da Cromatina , Impressão Genômica , Humanos , Neoplasias/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Inativação do Cromossomo X/genética
16.
Mol Cell Biol ; 29(6): 1389-400, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19114554

RESUMO

TFIIB recognizes DNA sequence-specific motifs that can flank the TATA elements of the promoters of protein-encoding genes. The TFIIB recognition elements (BRE(u) and BRE(d)) can have positive or negative effects on transcription in a promoter context-dependent manner. Here we show that the BREs direct the selective recruitment of TFIIA and NC2 to the promoter. We find that TFIIA preferentially associates with BRE-containing promoters while NC2 is recruited to promoters that lack consensus BREs. The functional relevance of the BRE-dependent recruitment of TFIIA and NC2 was determined by small interfering RNA-mediated knockdown of TFIIA and NC2, both of which elicited BRE-dependent effects on transcription. Our results confirm the established functional reciprocity of TFIIA and NC2. However, our findings show that TFIIA assembly at BRE-containing promoters results in reduced transcriptional activity, while NC2 acts as a positive factor at promoters that lack functional BREs. Taken together, our results provide a basis for the selective recruitment of TFIIA and NC2 to the promoter and give new insights into the functional relationship between core promoter elements and general transcription factor activity.


Assuntos
Fosfoproteínas/metabolismo , Fator de Transcrição TFIIA/metabolismo , Fator de Transcrição TFIIB/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular , Humanos , Fosfoproteínas/genética , Regiões Promotoras Genéticas , Fator de Transcrição TFIIA/genética , Fator de Transcrição TFIIB/genética , Fatores de Transcrição/genética , Transcrição Gênica
17.
J Biol Chem ; 282(34): 24767-76, 2007 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-17584739

RESUMO

Core promoter regions of protein-coding genes in metazoan genomes are structurally highly diverse and can contain several distinct core promoter elements, which direct accurate transcription initiation and determine basal promoter strength. Diversity in core promoter structure is an important aspect of transcription regulation in metazoans as it provides a basis for gene-selective function of activators and repressors. The basal activity of TATA box-containing promoters is dramatically enhanced by the initiator element (INR), which can function in concert with the TATA box in a synergistic manner. Here we report that a functional INR provides resistance to NC2 (Dr1/DRAP1), a general repressor of TATA promoters. INR-mediated resistance to NC2 is established during transcription initiation complex assembly and requires TBP-associated factors (TAFs) and TAF- and INR-dependent cofactor activity. Remarkably, the INR appears to stimulate TATA-dependent transcription similar to activators by strongly enhancing recruitment of TFIIA and TFIIB and, at the same time, by compromising NC2 binding.


Assuntos
Fosfoproteínas/fisiologia , TATA Box , Fatores de Transcrição/fisiologia , Transcrição Gênica , Núcleo Celular/metabolismo , HIV-1/genética , Células HeLa , Humanos , Modelos Biológicos , Modelos Genéticos , Fosfoproteínas/química , Regiões Promotoras Genéticas , Ligação Proteica , Fator de Transcrição TFIIA/metabolismo , Fator de Transcrição TFIIB/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
18.
J Biol Chem ; 280(52): 43087-99, 2005 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-16195225

RESUMO

Cytokine-dependent gene activation critically depends upon the tyrosine phosphorylation (activation) of STAT transcription factors at membrane-bound cytokine receptors. The extent of STAT activation and hence the specificity of signaling is primarily determined by structural complementarity between the SH2 domain of the STATs and the tyrosine-phosphorylated receptor chains. Here, we identified constitutive nucleocytoplasmic shuttling as another mechanism that controls the differential activation of STAT transcription factors. Our analysis of nucleocytoplasmic cycling of STAT1 revealed that the expression of the alternatively spliced transactivation domain and its signal-dependent serine phosphorylation maximized the rate of nuclear export. Export modulation occurred independently of retention factors or the export receptor CRM1, and was observed both before and during stimulation of cells with cytokines. Our data indicated a dual role for the transactivation domain. It enhanced the nuclear retention of activated STAT1, but had the opposite effect on inactivated molecules. Accordingly, and despite their identical receptor recognition, the STAT1 splice variants differed strongly in the amplitude of tyrosine phosphorylation and in the duration of the cytokine signal. Thus, regulated nuclear export determined the cytokine sensitivity of the shuttling STAT1 transcription factors by controlling their availability at the receptor kinase complex.


Assuntos
Transporte Ativo do Núcleo Celular , Citocinas/metabolismo , Fatores de Transcrição STAT/metabolismo , Processamento Alternativo , Western Blotting , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Dimerização , Relação Dose-Resposta a Droga , Genes Reporter , Glutationa Transferase/metabolismo , Proteínas de Fluorescência Verde/química , Células HeLa , Humanos , Microscopia Confocal , Modelos Biológicos , Peptídeos/química , Fosforilação , Plasmídeos/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Fator de Transcrição STAT1/metabolismo , Serina/química , Fatores de Tempo , Transcrição Gênica , Ativação Transcricional , Tirosina/química , Domínios de Homologia de src
19.
Genomics ; 80(3): 295-302, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12213199

RESUMO

We developed a novel efficient scheme, DEFOG (for "deciphering families of genes"), for determining sequences of numerous genes from a family of interest. The scheme provides a powerful means to obtain a gene family composition in species for which high-throughput genomic sequencing data are not available. DEFOG uses two key procedures. The first is a novel algorithm for designing highly degenerate primers based on a set of known genes from the family of interest. These primers are used in PCR reactions to amplify the members of the gene family. The second combines oligofingerprinting of the cloned PCR products with clustering of the clones based on their fingerprints. By selecting members from each cluster, a low-redundancy clone subset is chosen for sequencing. We applied the scheme to the human olfactory receptor (OR) genes. OR genes constitute the largest gene superfamily in the human genome, as well as in the genomes of other vertebrate species. DEFOG almost tripled the size of the initial repertoire of human ORs in a single experiment, and only 7% of the PCR clones had to be sequenced. Extremely high degeneracies, reaching over a billion combinations of distinct PCR primer pairs, proved to be very effective and yielded only 0.4% nonspecific products.


Assuntos
Família Multigênica , Análise de Sequência de DNA/métodos , Algoritmos , Sequência de Bases , Primers do DNA , Humanos , Receptores Odorantes/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA