Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Mol Cell ; 76(3): 453-472.e8, 2019 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-31519520

RESUMO

MYOD-directed fibroblast trans-differentiation into skeletal muscle provides a unique model to investigate how one transcription factor (TF) reconfigures the three-dimensional chromatin architecture to control gene expression, which is otherwise achieved by the combinatorial activities of multiple TFs. Integrative analysis of genome-wide high-resolution chromatin interactions, MYOD and CTCF DNA-binding profile, and gene expression, revealed that MYOD directs extensive re-wiring of interactions involving cis-regulatory and structural genomic elements, including promoters, enhancers, and insulated neighborhoods (INs). Re-configured INs were hot-spots of differential interactions, whereby MYOD binding to highly constrained sequences at IN boundaries and/or inside INs led to alterations of promoter-enhancer interactions to repress cell-of-origin genes and to activate muscle-specific genes. Functional evidence shows that MYOD-directed re-configuration of chromatin interactions temporally preceded the effect on gene expression and was mediated by direct MYOD-DNA binding. These data illustrate a model whereby a single TF alters multi-loop hubs to drive somatic cell trans-differentiation.


Assuntos
Transdiferenciação Celular , Reprogramação Celular , Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , Fibroblastos/metabolismo , Desenvolvimento Muscular , Proteína MyoD/metabolismo , Mioblastos Esqueléticos/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Transdiferenciação Celular/genética , Cromatina/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Desenvolvimento Muscular/genética , Proteína MyoD/genética , Conformação de Ácido Nucleico , Fenótipo , Ligação Proteica , Relação Estrutura-Atividade , Transcrição Gênica
2.
Nucleic Acids Res ; 45(10): 5739-5756, 2017 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-28334849

RESUMO

Hypomorphic mutations in DNA-methyltransferase DNMT3B cause majority of the rare disorder Immunodeficiency, Centromere instability and Facial anomalies syndrome cases (ICF1). By unspecified mechanisms, mutant-DNMT3B interferes with lymphoid-specific pathways resulting in immune response defects. Interestingly, recent findings report that DNMT3B shapes intragenic CpG-methylation of highly-transcribed genes. However, how the DNMT3B-dependent epigenetic network modulates transcription and whether ICF1-specific mutations impair this process remains unknown. We performed a transcriptomic and epigenomic study in patient-derived B-cell lines to investigate the genome-scale effects of DNMT3B dysfunction. We highlighted that altered intragenic CpG-methylation impairs multiple aspects of transcriptional regulation, like alternative TSS usage, antisense transcription and exon splicing. These defects preferentially associate with changes of intragenic H3K4me3 and at lesser extent of H3K27me3 and H3K36me3. In addition, we highlighted a novel DNMT3B activity in modulating the self-regulatory circuit of sense-antisense pairs and the exon skipping during alternative splicing, through interacting with RNA molecules. Strikingly, altered transcription affects disease relevant genes, as for instance the memory-B cell marker CD27 and PTPRC genes, providing us with biological insights into the ICF1-syndrome pathogenesis. Our genome-scale approach sheds light on the mechanisms still poorly understood of the intragenic function of DNMT3B and DNA methylation in gene expression regulation.


Assuntos
Processamento Alternativo , Anorexia/genética , Caquexia/genética , DNA (Citosina-5-)-Metiltransferases/genética , Anormalidades do Olho/genética , Histonas/genética , Síndromes de Imunodeficiência/genética , Mutação , RNA Mensageiro/genética , Dermatopatias/genética , Anorexia/imunologia , Anorexia/patologia , Linfócitos B/imunologia , Linfócitos B/patologia , Caquexia/imunologia , Caquexia/patologia , Linhagem Celular Transformada , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/imunologia , Metilação de DNA , Epigênese Genética , Anormalidades do Olho/imunologia , Anormalidades do Olho/patologia , Fácies , Feminino , Histonas/imunologia , Humanos , Síndromes de Imunodeficiência/imunologia , Síndromes de Imunodeficiência/patologia , Memória Imunológica , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/imunologia , Masculino , Regiões Promotoras Genéticas , RNA Mensageiro/imunologia , Dermatopatias/imunologia , Dermatopatias/patologia , Transcrição Gênica , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , DNA Metiltransferase 3B
3.
Mol Cancer Ther ; 20(6): 975-985, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33722854

RESUMO

KRASG12C inhibitors, including MRTX849, are promising treatment options for KRAS-mutant non-small cell lung cancer (NSCLC). PD-1 inhibitors are approved in NSCLC; however, strategies to enhance checkpoint inhibitor therapy (CIT) are needed. KRASG12C mutations are smoking-associated transversion mutations associated with high tumor mutation burden, PD-L1 positivity, and an immunosuppressive tumor microenvironment. To evaluate the potential of MRTX849 to augment CIT, its impact on immune signaling and response to CIT was evaluated. In human tumor xenograft models, MRTX849 increased MHC class I protein expression and decreased RNA and/or plasma protein levels of immunosuppressive factors. In a KrasG12C -mutant CT26 syngeneic mouse model, MRTX849 decreased intratumoral myeloid-derived suppressor cells and increased M1-polarized macrophages, dendritic cells, CD4+, and CD8+ T cells. Similar results were observed in lung KrasG12C -mutant syngeneic and a genetically engineered mouse (GEM) model. In the CT26 KrasG12C model, MRTX849 demonstrated marked tumor regression when tumors were established in immune-competent BALB/c mice; however, the effect was diminished when tumors were grown in T-cell-deficient nu/nu mice. Tumors progressed following anti-PD-1 or MRTX849 single-agent treatment in immune-competent mice; however, combination treatment demonstrated durable, complete responses (CRs). Tumors did not reestablish in the same mice that exhibited durable CRs when rechallenged with tumor cell inoculum, demonstrating these mice developed adaptive antitumor immunity. In a GEM model, treatment with MRTX849 plus anti-PD-1 led to increased progression-free survival compared with either single agent alone. These data demonstrate KRAS inhibition reverses an immunosuppressive tumor microenvironment and sensitizes tumors to CIT through multiple mechanisms.


Assuntos
Acetonitrilas/antagonistas & inibidores , Antineoplásicos/uso terapêutico , Inibidores de Checkpoint Imunológico/uso terapêutico , Piperazinas/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Pirimidinas/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Camundongos , Microambiente Tumoral/efeitos dos fármacos
4.
Cancer Discov ; 10(1): 54-71, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31658955

RESUMO

Despite decades of research, efforts to directly target KRAS have been challenging. MRTX849 was identified as a potent, selective, and covalent KRASG12C inhibitor that exhibits favorable drug-like properties, selectively modifies mutant cysteine 12 in GDP-bound KRASG12C, and inhibits KRAS-dependent signaling. MRTX849 demonstrated pronounced tumor regression in 17 of 26 (65%) KRASG12C-positive cell line- and patient-derived xenograft models from multiple tumor types, and objective responses have been observed in patients with KRASG12C-positive lung and colon adenocarcinomas. Comprehensive pharmacodynamic and pharmacogenomic profiling in sensitive and partially resistant nonclinical models identified mechanisms implicated in limiting antitumor activity including KRAS nucleotide cycling and pathways that induce feedback reactivation and/or bypass KRAS dependence. These factors included activation of receptor tyrosine kinases (RTK), bypass of KRAS dependence, and genetic dysregulation of cell cycle. Combinations of MRTX849 with agents that target RTKs, mTOR, or cell cycle demonstrated enhanced response and marked tumor regression in several tumor models, including MRTX849-refractory models. SIGNIFICANCE: The discovery of MRTX849 provides a long-awaited opportunity to selectively target KRASG12C in patients. The in-depth characterization of MRTX849 activity, elucidation of response and resistance mechanisms, and identification of effective combinations provide new insight toward KRAS dependence and the rational development of this class of agents.See related commentary by Klempner and Hata, p. 20.This article is highlighted in the In This Issue feature, p. 1.


Assuntos
Acetonitrilas/uso terapêutico , Adenocarcinoma de Pulmão/tratamento farmacológico , Antineoplásicos/uso terapêutico , Modelos Animais de Doenças , Neoplasias Pulmonares/tratamento farmacológico , Mutação , Piperazinas/uso terapêutico , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/genética , Pirrolidinas/uso terapêutico , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/patologia , Animais , Apoptose , Proliferação de Células , Ensaios Clínicos Fase I como Assunto , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Pessoa de Meia-Idade , Prognóstico , Pirimidinas , Transdução de Sinais , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Nat Commun ; 10(1): 4148, 2019 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31515477

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD), caused by mutations in either PKD1 or PKD2 genes, is one of the most common human monogenetic disorders and the leading genetic cause of end-stage renal disease. Unfortunately, treatment options for ADPKD are limited. Here we report the discovery and characterization of RGLS4326, a first-in-class, short oligonucleotide inhibitor of microRNA-17 (miR-17), as a potential treatment for ADPKD. RGLS4326 is discovered by screening a chemically diverse and rationally designed library of anti-miR-17 oligonucleotides for optimal pharmaceutical properties. RGLS4326 preferentially distributes to kidney and collecting duct-derived cysts, displaces miR-17 from translationally active polysomes, and de-represses multiple miR-17 mRNA targets including Pkd1 and Pkd2. Importantly, RGLS4326 demonstrates a favorable preclinical safety profile and attenuates cyst growth in human in vitro ADPKD models and multiple PKD mouse models after subcutaneous administration. The preclinical characteristics of RGLS4326 support its clinical development as a disease-modifying treatment for ADPKD.


Assuntos
MicroRNAs/antagonistas & inibidores , Oligonucleotídeos/uso terapêutico , Doenças Renais Policísticas/tratamento farmacológico , Doenças Renais Policísticas/genética , Animais , Sequência de Bases , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Redes Reguladoras de Genes/efeitos dos fármacos , Células HeLa , Hematopoese/efeitos dos fármacos , Humanos , Túbulos Renais/patologia , Macaca fascicularis , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Oligonucleotídeos/farmacocinética , Oligonucleotídeos/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Distribuição Tecidual/efeitos dos fármacos
7.
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
8.
Nat Cell Biol ; 20(8): 917-927, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30050118

RESUMO

Fibro-adipogenic progenitors (FAPs) are typically activated in response to muscle injury, and establish functional interactions with inflammatory and muscle stem cells (MuSCs) to promote muscle repair. We found that denervation causes progressive accumulation of FAPs, without concomitant infiltration of macrophages and MuSC-mediated regeneration. Denervation-activated FAPs exhibited persistent STAT3 activation and secreted elevated levels of IL-6, which promoted muscle atrophy and fibrosis. FAPs with aberrant activation of STAT3-IL-6 signalling were also found in mouse models of spinal cord injury, spinal muscular atrophy, amyotrophic lateral sclerosis (ALS) and in muscles of ALS patients. Inactivation of STAT3-IL-6 signalling in FAPs effectively countered muscle atrophy and fibrosis in mouse models of acute denervation and ALS (SODG93A mice). Activation of pathogenic FAPs following loss of integrity of neuromuscular junctions further illustrates the functional versatility of FAPs in response to homeostatic perturbations and suggests their potential contribution to the pathogenesis of neuromuscular diseases.


Assuntos
Adipogenia , Esclerose Lateral Amiotrófica/metabolismo , Denervação/métodos , Interleucina-6/metabolismo , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular/metabolismo , Mioblastos Esqueléticos/metabolismo , Músculo Quadríceps/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Traumatismos da Medula Espinal/metabolismo , Adipogenia/efeitos dos fármacos , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/prevenção & controle , Animais , Cardiotoxinas , Linhagem Celular , Técnicas de Cocultura , Modelos Animais de Doenças , Fibrose , Humanos , Interleucina-6/antagonistas & inibidores , Interleucina-6/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atrofia Muscular/genética , Atrofia Muscular/patologia , Atrofia Muscular/prevenção & controle , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patologia , Atrofia Muscular Espinal/prevenção & controle , Mutação , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/patologia , Fármacos Neuromusculares/farmacologia , Músculo Quadríceps/efeitos dos fármacos , Músculo Quadríceps/inervação , Músculo Quadríceps/patologia , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Nervo Isquiático/cirurgia , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/prevenção & controle , Superóxido Dismutase-1/genética
9.
Mol Ther Nucleic Acids ; 13: 1-15, 2018 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-30219269

RESUMO

Extracellular small RNAs (sRNAs), including microRNAs (miRNAs), are promising biomarkers for diseases such as Duchenne muscular dystrophy (DMD), although their biological relevance is largely unknown. To investigate the relationship between intracellular and extracellular sRNA levels on a global scale, we performed sRNA sequencing in four muscle types and serum from wild-type, dystrophic mdx, and mdx mice in which dystrophin protein expression was restored by exon skipping. Differentially abundant sRNAs were identified in serum (mapping to miRNA, small nuclear RNA [snRNA], and PIWI-interacting RNA [piRNA] loci). One novel candidate biomarker, miR-483, was increased in both mdx serum and muscle, and also elevated in DMD patient sera. Dystrophin restoration induced global shifts in miRNA (including miR-483) and snRNA-fragment abundance toward wild-type levels. Specific serum piRNA-like sRNAs also responded to exon skipping therapy. Absolute miRNA expression in muscle was positively correlated with abundance in the circulation, although multiple highly expressed miRNAs in muscle were not elevated in mdx serum, suggesting that both passive and selective release mechanisms contribute to serum miRNA levels. In conclusion, this study has revealed new insights into the sRNA biology of dystrophin deficiency and identified novel DMD biomarkers.

10.
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
11.
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
12.
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
13.
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
14.
Epigenetics ; 7(6): 542-50, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22595875

RESUMO

The immunodeficiency, centromere instability and facial anomalies (ICF) syndrome is associated to mutations of the DNA methyl-transferase DNMT3B, resulting in a reduction of enzyme activity. Aberrant expression of immune system genes and hypomethylation of pericentromeric regions accompanied by chromosomal instability were determined as alterations driving the disease phenotype. However, so far only technologies capable to analyze single loci were applied to determine epigenetic alterations in ICF patients. In the current study, we performed whole-genome bisulphite sequencing to assess alteration in DNA methylation at base pair resolution. Genome-wide we detected a decrease of methylation level of 42%, with the most profound changes occurring in inactive heterochromatic regions, satellite repeats and transposons. Interestingly, transcriptional active loci and ribosomal RNA repeats escaped global hypomethylation. Despite a genome-wide loss of DNA methylation the epigenetic landscape and crucial regulatory structures were conserved. Remarkably, we revealed a mislocated activity of mutant DNMT3B to H3K4me1 loci resulting in hypermethylation of active promoters. Functionally, we could associate alterations in promoter methylation with the ICF syndrome immunodeficient phenotype by detecting changes in genes related to the B-cell receptor mediated maturation pathway.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , Síndromes de Imunodeficiência/genética , Linfócitos B , Linhagem Celular Transformada , Pré-Escolar , DNA (Citosina-5-)-Metiltransferases/metabolismo , Epigênese Genética , Face/anormalidades , Feminino , Genoma Humano , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Síndromes de Imunodeficiência/metabolismo , Mutação , Doenças da Imunodeficiência Primária , Análise de Sequência de DNA , Sulfitos , DNA Metiltransferase 3B
15.
Epigenetics ; 6(10): 1242-7, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21931280

RESUMO

Molecular mechanisms underlying aberrant phenotypes in balanced X;autosome translocations are scarcely understood. We report the case of a de novo reciprocal balanced translocation X;2(q23;q33) presenting phenotypic alterations highly suggestive of Incontinentia Pigmenti (IP) syndrome, a genodermatosis with abnormal skin pigmentation and neurological failure, segregating as X-linked dominant disorder. Through molecular studies, we demonstrated that the altered phenotype could not be ascribed to chromosome microdeletions or to XIST-mediated inactivation of Xq24-qter. Interestingly, we found that the Xq24-qter region, which translocated downstream of the heterochromatic band 2q34, undergoes epigenetic silencing mediated by DNA methylation and histone alterations. Among the downregulated genes, we found the inhibitor of kappa light polypeptide gene enhancer in B cells, kinase gamma (IKBKG/NEMO), the causative gene of IP. We hypothesize that a mosaic functional nullisomy of the translocated genes, through a Position Effect Variegation-like heterochromatization, might be responsible for the proband's phenotypic anomalies. Partial silencing of IKBKG may be responsible for the skin anomalies observed, thereby mimicking the IP pathological condition. In addition to its clinical relevance, this paper addresses fundamental issues related to the chromatin status and nuclear localization of a human euchromatic region translocated proximally to heterochromatin. In conclusion, the study provides new insight into long-range gene silencing mechanisms and their direct impact in human disease.


Assuntos
Cromossomos Humanos X , Epigênese Genética , Inativação Gênica , Incontinência Pigmentar/genética , Metilação de DNA , Histonas/metabolismo , Humanos , Quinase I-kappa B/genética , Fenótipo , Translocação Genética
16.
Epigenetics ; 5(5): 427-43, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20448464

RESUMO

Immunodeficiency, Centromeric region instability, Facial anomalies (ICF; OMIM #242860) syndrome, due to mutations in the DNMT3B gene, is characterized by inheritance of aberrant patterns of DNA methylation and heterochromatin defects. Patients show variable agammaglobulinemia and a reduced number of T cells, making them prone to infections and death before adulthood. Other variable symptoms include facial dysmorphism, growth and mental retardation. Despite the recent advances in identifying the dysregulated genes, the molecular mechanisms, which underlie the altered gene expression causing ICF phenotype complexity, are not well understood. Held the recently-shown tight correlation between epigenetics and microRNAs (miRNAs), we searched for miRNAs regulated by DNMT3B activity, comparing cell lines from ICF patients with those from healthy individuals. We observe that eighty-nine miRNAs, some of which involved in immune function, development and neurogenesis, are dysregulated in ICF (LCLs) compared to wild-type cells. Significant DNA hypomethylation of miRNA CpG islands was not observed in cases of miRNA up-regulation in ICF cells, suggesting a more subtle effect of DNMT3B deficiency on their regulation; however, a modification of histone marks, especially H3K27 and H3K4 trimethylation, and H4 acetylation, was observed concomitantly with changes in microRNA expression. Functional correlation between miRNA and mRNA expression of their targets allow us to suppose a regulation either at mRNA level or at protein level. These results provide a better understanding of how DNA methylation and histone code interact to regulate the class of microRNA genes and enable us to predict molecular events possibly contributing to ICF condition.


Assuntos
Anormalidades Múltiplas/genética , Instabilidade Cromossômica/genética , DNA (Citosina-5-)-Metiltransferases/genética , Epigênese Genética/genética , MicroRNAs/genética , Mutação , Anormalidades Múltiplas/enzimologia , Ilhas de CpG , Face/anormalidades , Histonas/genética , Histonas/metabolismo , Humanos , Síndromes de Imunodeficiência/enzimologia , Síndromes de Imunodeficiência/genética , MicroRNAs/metabolismo , Doenças da Imunodeficiência Primária , Regulação para Cima , DNA Metiltransferase 3B
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA