Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Sci Adv ; 10(18): eadj8042, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38691608

RESUMEN

Overactivation of the transforming growth factor-ß (TGFß) signaling in Duchenne muscular dystrophy (DMD) is a major hallmark of disease progression, leading to fibrosis and muscle dysfunction. Here, we investigated the role of SETDB1 (SET domain, bifurcated 1), a histone lysine methyltransferase involved in muscle differentiation. Our data show that, following TGFß induction, SETDB1 accumulates in the nuclei of healthy myotubes while being already present in the nuclei of DMD myotubes where TGFß signaling is constitutively activated. Transcriptomics revealed that depletion of SETDB1 in DMD myotubes leads to down-regulation of TGFß target genes coding for secreted factors involved in extracellular matrix remodeling and inflammation. Consequently, SETDB1 silencing in DMD myotubes abrogates the deleterious effect of their secretome on myoblast differentiation by impairing myoblast pro-fibrotic response. Our findings indicate that SETDB1 potentiates the TGFß-driven fibrotic response in DMD muscles, providing an additional axis for therapeutic intervention.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Fibras Musculares Esqueléticas , Distrofia Muscular de Duchenne , Transducción de Señal , Factor de Crecimiento Transformador beta , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Factor de Crecimiento Transformador beta/metabolismo , Humanos , Animales , Diferenciación Celular , Ratones , Mioblastos/metabolismo , Fibrosis , Regulación de la Expresión Génica
2.
iScience ; 26(8): 107386, 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37559904

RESUMEN

The major lysine methyltransferase (KMT) Setdb1 is essential for self-renewal and viability of mouse embryonic stem cells (mESCs). Setdb1 was primarily known to methylate the lysine 9 of histone 3 (H3K9) in the nucleus, where it regulates chromatin functions. However, Setdb1 is also massively localized in the cytoplasm, including in mESCs, where its role remains elusive. Here, we show that the cytoplasmic Setdb1 (cSetdb1) is essential for the survival of mESCs. Yeast two-hybrid analysis revealed that cSetdb1 interacts with several regulators of mRNA stability and protein translation machinery, such as the ESCs-specific E3 ubiquitin ligase and mRNA silencer Trim71/Lin41. We found that cSetdb1 is required for the integrity of Trim71 complex(es) involved in mRNA metabolism and translation. cSetdb1 modulates the abundance of mRNAs and the rate of newly synthesized proteins. Altogether, our data uncovered the cytoplasmic post-transcriptional regulation of gene expression mediated by a key epigenetic regulator.

3.
Int J Mol Sci ; 24(14)2023 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-37511179

RESUMEN

Duchenne muscular dystrophy (DMD) is a yet incurable rare genetic disease that affects the skeletal and cardiac muscles, leading to progressive muscle wasting and premature death. DMD is caused by the lack of dystrophin, a muscle protein essential for the biochemical support and integrity of muscle fibers. Gene replacement strategies for Duchenne muscular dystrophy (DMD) employing the adeno-associated virus (AAV) face the challenge imposed by the limited packaging capacity of AAV, only allowing the accommodation of a short version of dystrophin (µDys) that is still far removed from correcting human disease. The need to develop strategies leading to the expression of a best performing dystrophin variant led to only few studies reporting on the use of dual vectors, but none reported on a method to assess in vivo transgene reconstitution efficiency, the degree of which directly affects the use of safe AAV dosing. We report here on the generation of a dual AAV vector approach for the expression of a larger dystrophin version (quasidystrophin) based on homologous recombination, and the development of a methodology employing a strategic droplet digital PCR design, to determine the recombination efficiency as well as the occurrence of unwanted concatemerization events or aberrant expression from the single vectors. We demonstrated that, upon systemic delivery in the dystrophic D2.B10-Dmdmdx/J (DBA2mdx) mice, our dual AAV approach led to high transgene reconstitution efficiency and negligible Inverted Terminal Repeats (ITR)-dependent concatemerization, with consequent remarkable protein restoration in muscles and improvement of muscle pathology. This evidence supports the suitability of our system for gene therapy application and the potential of this methodology to assess and improve the feasibility for therapeutic translation of multiple vector approaches.


Asunto(s)
Distrofia Muscular de Duchenne , Ratones , Humanos , Animales , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/metabolismo , Distrofina/genética , Distrofina/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Músculo Esquelético/metabolismo , Ratones Endogámicos mdx , Vectores Genéticos/genética
4.
Nat Commun ; 14(1): 292, 2023 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-36653361

RESUMEN

Pancreatic cancer is characterized by extensive resistance to conventional therapies, making clinical management a challenge. Here we map the epigenetic dependencies of cancer stem cells, cells that preferentially evade therapy and drive progression, and identify SWI/SNF complex member SMARCD3 as a regulator of pancreatic cancer cells. Although SWI/SNF subunits often act as tumor suppressors, we show that SMARCD3 is amplified in cancer, enriched in pancreatic cancer stem cells and upregulated in the human disease. Diverse genetic mouse models of pancreatic cancer and stage-specific Smarcd3 deletion reveal that Smarcd3 loss preferentially impacts established tumors, improving survival especially in context of chemotherapy. Mechanistically, SMARCD3 acts with FOXA1 to control lipid and fatty acid metabolism, programs associated with therapy resistance and poor prognosis in cancer. These data identify SMARCD3 as an epigenetic modulator responsible for establishing the metabolic landscape in aggressive pancreatic cancer cells and a potential target for new therapies.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Ratones , Animales , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Epigénesis Genética , Neoplasias Pancreáticas
5.
Biochim Biophys Acta Proteins Proteom ; 1869(5): 140616, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33545367

RESUMEN

This review was written in memory of our late friend, Dr. Hiroyuki Sorimachi, who, following the steps of his mentor Koichi Suzuki, a pioneer in calpain research, has made tremendous contributions to the field. During his career, Hiro also wrote several reviews on calpain, the last of which, published in 2016, was comprehensive. In this manuscript, we decided to put together a review with the basic information a novice may need to know about calpains. We also tried to avoid similarities with previous reviews and reported the most significant new findings, at the same time highlighting Hiro's contributions to the field. The review will cover a short history of calpain discovery, the presentation of the family, the life of calpain from transcription to activity, human diseases caused by calpain mutations and therapeutic perspectives.


Asunto(s)
Calpaína/fisiología , Calpaína/genética , Calpaína/metabolismo , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/terapia , Terapia Genética , Humanos , Lisosomas/metabolismo , Mutación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis
6.
Skelet Muscle ; 10(1): 13, 2020 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-32359374

RESUMEN

BACKGROUND: In Duchenne muscular dystrophy (DMD), DYSTROPHIN deficiency exposes myofibers to repeated cycles of contraction/degeneration, ultimately leading to muscle loss and replacement by fibrotic tissue. DMD pathology is typically exacerbated by excessive secretion of TGFß and consequent accumulation of pro-fibrotic components of the extra-cellular matrix (ECM), which in turn impairs compensatory regeneration and complicates the efficacy of therapeutic strategies. It is currently unclear whether DMD skeletal muscle fibers directly contribute to excessive activation of TGFß. Development of skeletal myofibers from DMD patient-derived induced pluripotent stem cells (iPSC), as an "in dish" model of disease, can be exploited to determine the myofiber contribution to pathogenic TGFß signaling in DMD and might provide a screening platform for the identification of anti-fibrotic interventions in DMD. METHODS: We describe a rapid and efficient method for the generation of contractile human skeletal muscle cells from DMD patient-derived hiPSC, based on the inducible expression of MyoD and BAF60C (encoded by SMARCD3 gene), using an enhanced version of piggyBac (epB) transposone vectors. DMD iPSC-derived myotubes were tested as an "in dish" disease model and exposed to environmental and mechanical cues that recapitulate salient pathological features of DMD. RESULTS: We show that DMD iPSC-derived myotubes exhibit a constitutive activation of TGFß-SMAD2/3 signaling. High-content screening (HCS)-based quantification of nuclear phosphorylated SMAD2/3 signal revealed that DMD iPSC-derived myotubes also exhibit increased activation of the TGFß-SMAD2/3 signaling following exposure to either recombinant TGFß or electrical pacing-induced contraction. CONCLUSIONS: Acute conversion of DMD patient-derived iPSC into skeletal muscles, by the ectopic expression of MyoD and BAF60C, provides a rapid and reliable protocol for an "in dish" DMD model that recapitulates key pathogenic features of disease pathology, such as the constitutive activation of the TGFß/SMAD signaling as well as the deregulated response to pathogenic stimuli, e.g., ECM-derived signals or mechanical cues. Thus, this model is suitable for the identification of new therapeutic targets in DMD patient-specific muscles.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Cultivo Primario de Células/métodos , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Diferenciación Celular , Línea Celular , Células Cultivadas , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Fibrosis , Humanos , Células Madre Pluripotentes Inducidas/citología , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Proteína MioD/genética , Proteína MioD/metabolismo , Proteínas Smad/genética , Proteínas Smad/metabolismo
7.
Genes Dev ; 31(13): 1325-1338, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28794185

RESUMEN

Deciphering the fundamental mechanisms controlling cardiac specification is critical for our understanding of how heart formation is initiated during embryonic development and for applying stem cell biology to regenerative medicine and disease modeling. Using systematic and unbiased functional screening approaches, we discovered that the Id family of helix-loop-helix proteins is both necessary and sufficient to direct cardiac mesoderm formation in frog embryos and human embryonic stem cells. Mechanistically, Id proteins specify cardiac cell fate by repressing two inhibitors of cardiogenic mesoderm formation-Tcf3 and Foxa2-and activating inducers Evx1, Grrp1, and Mesp1. Most importantly, CRISPR/Cas9-mediated ablation of the entire Id (Id1-4) family in mouse embryos leads to failure of anterior cardiac progenitor specification and the development of heartless embryos. Thus, Id proteins play a central and evolutionarily conserved role during heart formation and provide a novel means to efficiently produce cardiovascular progenitors for regenerative medicine and drug discovery applications.


Asunto(s)
Linaje de la Célula/genética , Corazón/embriología , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Organogénesis/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Línea Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión no Mamífero/citología , Embrión no Mamífero/embriología , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Edición Génica , Regulación del Desarrollo de la Expresión Génica/genética , Cardiopatías Congénitas/genética , Humanos , Mesodermo/citología , Mesodermo/fisiología , Ratones , Mutación , Semillas , Xenopus laevis/embriología
8.
Cell Mol Life Sci ; 73(20): 3887-96, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27207468

RESUMEN

SWI/SNF chromatin-remodeling complexes are key regulators of the epigenetic modifications that determine whether stem cells maintain pluripotency or commit toward specific lineages through development and during postnatal life. Dynamic combinatorial assembly of multiple variants of SWI/SNF subunits is emerging as the major determinant of the functional versatility of SWI/SNF. Here, we summarize the current knowledge on the structural and functional properties of the alternative SWI/SNF complexes that direct stem cell fate toward skeletal muscle lineage and control distinct stages of skeletal myogenesis. In particular, we will refer to recent evidence pointing to the essential role of two SWI/SNF components not expressed in embryonic stem cells-the catalytic subunit BRM and the structural component BAF60C-whose induction in muscle progenitors coincides with the expansion of their transcriptional repertoire.


Asunto(s)
Linaje de la Célula , Proteínas Cromosómicas no Histona/metabolismo , Desarrollo de Músculos , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Linaje de la Célula/genética , Humanos , Modelos Biológicos , Desarrollo de Músculos/genética
9.
Elife ; 52016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26880551

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica , Células Musculares/fisiología , Proteína MioD/metabolismo , Proteína de Unión a TATA-Box/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Ratones , Ratones Noqueados
10.
EMBO Rep ; 16(8): 1037-50, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26136374

RESUMEN

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.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , ADN Helicasas/metabolismo , Expresión Génica , Desarrollo de Músculos/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Ciclina D1/deficiencia , Ciclina D1/genética , ADN Helicasas/genética , Técnicas de Silenciamiento del Gen , Ratones , Células Musculares , Proteínas Nucleares/genética , Factores de Transcripción/genética
11.
J Vis Exp ; (88): e51243, 2014 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-24999032

RESUMEN

Generation of a homogeneous and abundant population of skeletal muscle cells from human embryonic stem cells (hESCs) is a requirement for cell-based therapies and for a "disease in a dish" model of human neuromuscular diseases. Major hurdles, such as low abundance and heterogeneity of the population of interest, as well as a lack of protocols for the formation of three-dimensional contractile structures, have limited the applications of stem cells for neuromuscular disorders. We have designed a protocol that overcomes these limits by ectopic introduction of defined factors in hESCs--the muscle determination factor MyoD and SWI/SNF chromatin remodeling complex component BAF60C--that are able to reprogram hESCs into skeletal muscle cells. Here we describe the protocol established to generate hESC-derived myoblasts and promote their clustering into tridimensional miniaturized structures (myospheres) that functionally mimic miniaturized skeletal muscles.


Asunto(s)
Bioingeniería/métodos , Células Madre Embrionarias/fisiología , Músculo Esquelético/fisiología , Proteínas Cromosómicas no Histona , Células Madre Embrionarias/citología , Epigenómica , Humanos , Músculo Esquelético/citología , Proteína MioD/genética , Factores de Transcripción/genética
12.
Genes Dev ; 27(21): 2332-44, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24186978

RESUMEN

A critical but molecularly uncharacterized step in heart formation and regeneration is the process that commits progenitor cells to differentiate into cardiomyocytes. Here, we show that the endoderm-derived dual Nodal/bone morphogenetic protein (BMP) antagonist Cerberus-1 (Cer1) in embryonic stem cell cultures orchestrates two signaling pathways that direct the SWI/SNF chromatin remodeling complex to cardiomyogenic loci in multipotent (KDR/Flk1+) progenitors, activating lineage-specific transcription. Transient inhibition of Nodal by Cer1 induces Brahma-associated factor 60c (Baf60c), one of three Baf60 variants (a, b, and c) that are mutually exclusively assembled into SWI/SNF. Blocking Nodal and BMP also induces lineage-specific transcription factors Gata4 and Tbx5, which interact with Baf60c. siRNA to Cer1, Baf60c, or the catalytic SWI/SNF subunit Brg1 prevented the developmental opening of chromatin surrounding the Nkx2.5 early cardiac enhancer and cardiomyocyte differentiation. Overexpression of Baf60c fully rescued these deficits, positioning Baf60c and SWI/SNF function downstream from Cer1. Thus, antagonism of Nodal and BMP coordinates induction of the myogenic Baf60c variant and interacting transcription factors to program the developmental opening of cardiomyocyte-specific loci in chromatin. This is the first demonstration that cues from the progenitor cell environment direct the subunit variant composition of SWI/SNF to remodel the transcriptional landscape for lineage-specific differentiation.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Miocitos Cardíacos/citología , Proteína Nodal/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Células Cultivadas , Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona , Citocinas/genética , Citocinas/metabolismo , Endodermo/metabolismo , Perfilación de la Expresión Génica , Humanos , Ratones , Miocitos Cardíacos/metabolismo , Proteína Nodal/genética , ARN Interferente Pequeño/metabolismo , Células Madre/citología , Células Madre/metabolismo
13.
Cell Rep ; 3(3): 661-70, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23478022

RESUMEN

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.


Asunto(s)
Reprogramación Celular , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Fibras Musculares Esqueléticas/citología , Línea Celular , Linaje de la Célula , Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona , Células Madre Embrionarias/citología , Humanos , Mesodermo/citología , Contracción Muscular , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/fisiología , Proteína MioD/genética , Proteína MioD/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
EMBO J ; 31(2): 301-16, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22068056

RESUMEN

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.


Asunto(s)
Proteínas Cromosómicas no Histona/fisiología , Sistema de Señalización de MAP Quinasas , Desarrollo de Músculos/fisiología , Proteínas Musculares/fisiología , Proteína MioD/fisiología , Factores de Transcripción/fisiología , Animales , Línea Celular , Cromatina/genética , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , ADN Helicasas/fisiología , Fibroblastos/metabolismo , Regulación de la Expresión Génica/genética , Células HeLa/metabolismo , Humanos , Ratones , Complejos Multiproteicos , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/química , Proteínas Musculares/genética , Mioblastos/metabolismo , Proteínas Nucleares/fisiología , Fosforilación , Fosfotreonina/análisis , Procesamiento Proteico-Postraduccional , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/química , Factores de Transcripción/genética , Técnicas del Sistema de Dos Híbridos , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
15.
Exp Cell Res ; 316(18): 3073-80, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20553711

RESUMEN

Skeletal muscle differentiation relies on the coordinated activation and repression of specific subsets of genes. This reflects extensive changes in chromatin architecture, composition of chromatin-associated complexes and histone modifications at the promoter/enhancer elements of skeletal muscle genes. An early, key event in the activation of muscle-specific gene transcription is the disruption of the repressive conformation imposed by nucleosomes, which impede the access of pioneer transcription factors, such as the muscle-specific basic helix-loop-helix (bHLH) factors MyoD and Myf5, to their DNA-binding sites. This review focuses on our current understanding of the role of the SWI/SNF ATP-dependent chromatin-remodeling complex in the activation of the myogenic program, by inducing conformational changes permissive for muscle-gene expression. Recent findings suggest that specific combinations of individual SWI/SNF components can generate sub-complexes with specialized functions that are engaged at sequential stages of muscle-gene activation--e.g., initial displacement of the nucleosome followed by the loading of the complete myogenic transcriptosome that promotes gene transcription. SWI/SNF composition and function is regulated by the exchange of specific variants of structural sub-units. In turn, an exchange of histone variants and related epigenetic modifications might reflect the impact of distinct SWI/SNF complexes on the architecture and activity of target promoter/enhancer elements. Thus, the SWI/SNF complexes should be regarded not just as simple executors of the program imposed by transcription factors, but as multifaceted "readers" and "shapers" of the chromatin/DNA landscape within target muscle genes along the transition from myoblasts to myotubes.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Proteínas Cromosómicas no Histona/fisiología , Desarrollo de Músculos/fisiología , Músculo Esquelético/citología , Factores de Transcripción/fisiología , Animales , Humanos
16.
Cancer Res ; 70(3): 916-24, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20103633

RESUMEN

Neuroblastoma (NB) is the most common solid extracranial cancer of childhood. Amplification and overexpression of the MYCN oncogene characterize the most aggressive forms and are believed to severely downregulate MHC class I molecules by transcriptional inhibition of the p50 NF-kappaB subunit. In this study, we found that in human NB cell lines, high MYCN expression is not responsible for low MHC class I expression because neither transfection-mediated overexpression nor small interfering RNA suppression of MYCN affects MHC class I and p50 levels. Furthermore, we identified NF-kappaB as the immediate upstream regulator of MHC class I because the p65 NF-kappaB subunit binds MHC class I promoter in chromatin immunoprecipitation experiments, and MHC class I expression is enhanced by p65 transfection and reduced by (a) the chemical NF-kappaB inhibitor sulfasalazine, (b) a dominant-negative IKBalpha gene, and (c) p65 silencing. Moreover, we showed that the endoplasmic reticulum aminopeptidases ERAP1 and ERAP2, which generate MHC class I binding peptides, are regulated by NF-kappaB, contain functional NF-kappaB-binding elements in their promoters, and mimic MHC class I molecules in the expression pattern. Consistent with these findings, nuclear p65 was detected in NB cells that express MHC class I molecules in human NB specimens. Thus, the coordinated downregulation of MHC class I, ERAP1, and ERAP2 in aggressive NB cells is attributable to a low transcriptional availability of NF-kappaB, possibly due to an unknown suppressor other than MYCN.


Asunto(s)
Aminopeptidasas/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , FN-kappa B/metabolismo , Aminopeptidasas/genética , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Antígenos de Histocompatibilidad Menor , Proteína Proto-Oncogénica N-Myc , FN-kappa B/genética , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Transporte de Proteínas/efectos de los fármacos , Interferencia de ARN , Transfección , Factor de Necrosis Tumoral alfa/farmacología
17.
Dev Cell ; 17(6): 836-48, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20059953

RESUMEN

We created a whole-mount in situ hybridization (WISH) database, termed EMBRYS, containing expression data of 1520 transcription factors and cofactors expressed in E9.5, E10.5, and E11.5 mouse embryos--a highly dynamic stage of skeletal myogenesis. This approach implicated 43 genes in regulation of embryonic myogenesis, including a transcriptional repressor, the zinc-finger protein RP58 (also known as Zfp238). Knockout and knockdown approaches confirmed an essential role for RP58 in skeletal myogenesis. Cell-based high-throughput transfection screening revealed that RP58 is a direct MyoD target. Microarray analysis identified two inhibitors of skeletal myogenesis, Id2 and Id3, as targets for RP58-mediated repression. Consistently, MyoD-dependent activation of the myogenic program is impaired in RP58 null fibroblasts and downregulation of Id2 and Id3 rescues MyoD's ability to promote myogenesis in these cells. Our combined, multi-system approach reveals a MyoD-activated regulatory loop relying on RP58-mediated repression of muscle regulatory factor (MRF) inhibitors.


Asunto(s)
Redes Reguladoras de Genes , Desarrollo de Músculos , Músculo Esquelético/embriología , Proteínas Represoras/metabolismo , Animales , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Proteínas Inhibidoras de la Diferenciación/metabolismo , Ratones , Factores Reguladores Miogénicos/genética
18.
PLoS One ; 3(5): e2236, 2008 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-18493594

RESUMEN

We identified a key oncogenic pathway underlying neuroblastoma progression: specifically, MYCN, expressed at elevated level, transactivates the miRNA 17-5p-92 cluster, which inhibits p21 and BIM translation by interaction with their mRNA 3' UTRs. Overexpression of miRNA 17-5p-92 cluster in MYCN-not-amplified neuroblastoma cells strongly augments their in vitro and in vivo tumorigenesis. In vitro or in vivo treatment with antagomir-17-5p abolishes the growth of MYCN-amplified and therapy-resistant neuroblastoma through p21 and BIM upmodulation, leading to cell cycling blockade and activation of apoptosis, respectively. In primary neuroblastoma, the majority of cases show a rise of miR-17-5p level leading to p21 downmodulation, which is particularly severe in patients with MYCN amplification and poor prognosis. Altogether, our studies demonstrate for the first time that antagomir treatment can abolish tumor growth in vivo, specifically in therapy-resistant neuroblastoma.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de la Membrana/metabolismo , MicroARNs/uso terapéutico , Neuroblastoma/patología , Proteína Oncogénica p21(ras)/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Secuencia de Bases , Proteína 11 Similar a Bcl2 , Línea Celular Tumoral , Regulación hacia Abajo , Resistencia a Antineoplásicos , Genes myc , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , MicroARNs/genética , Neuroblastoma/tratamiento farmacológico , ARN Interferente Pequeño
19.
Mol Cell Biol ; 27(20): 7248-65, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17709384

RESUMEN

The expression of retinoblastoma (pRb) and cyclin D3 proteins is highly induced during the process of skeletal myoblast differentiation. We have previously shown that cyclin D3 is nearly totally associated with hypophosphorylated pRb in differentiated myotubes, whereas Rb-/- myocytes fail to accumulate the cyclin D3 protein despite normal induction of cyclin D3 mRNA. Here we report that pRb promotes cyclin D3 protein accumulation in differentiating myoblasts by preventing cyclin D3 degradation. We show that cyclin D3 displays rapid turnover in proliferating myoblasts, which is positively regulated through glycogen synthase kinase 3beta (GSK-3beta)-mediated phosphorylation of cyclin D3 on Thr-283. We describe a novel interaction between pRb and cyclin D3 that maps to the C terminus of pRb and to a region of cyclin D3 proximal to the Thr-283 residue and provide evidence that the pRb-cyclin D3 complex formation in terminally differentiated myotubes hinders the access of GSK-3beta to cyclin D3, thus inhibiting Thr-283 phosphorylation. Interestingly, we observed that the ectopic expression of a stabilized cyclin D3 mutant in C2 myoblasts enhances muscle-specific gene expression; conversely, cyclin D3-null embryonic fibroblasts display impaired MyoD-induced myogenic differentiation. These results indicate that the pRb-dependent accumulation of cyclin D3 is functionally relevant to the process of skeletal muscle cell differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Ciclinas/metabolismo , Desarrollo de Músculos/fisiología , Proteína de Retinoblastoma/metabolismo , Animales , Células Cultivadas , Ciclina D3 , Ciclinas/genética , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Ratones , Ratones Noqueados , Mioblastos/citología , Mioblastos/fisiología , Fosforilación , Proteína de Retinoblastoma/genética , Treonina/metabolismo
20.
Clin Cancer Res ; 11(7): 2756-67, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15814658

RESUMEN

PURPOSE: Melanoma patients have a very poor prognosis with a response rate of <1% due to advanced diagnosis. This type of tumor is particularly resistant to conventional chemotherapy and radiotherapy, and the surgery remains the principal treatment for patients with localized melanoma. For this reason, there is particular interest in the melanoma biological therapy. EXPERIMENTAL DESIGN: Using two p53 mutant melanoma models stably expressing an inducible c-myc antisense RNA, we have investigated whether Myc protein down-regulation could render melanoma cells more susceptible to radiotherapy, reestablishing apoptotic p53-independent pathway. In addition to address the role of p53 in the activation of apoptosis, we studied the effect of Myc down-regulation on radiotherapy sensitivity also in a p53 wild-type melanoma cell line. RESULTS: Myc down-regulation is able per se to induce apoptosis in a fraction of the cell population (approximately 40% at 72 hours) and in combination with gamma radiation efficiently enhances the death process. In fact, approximately 80% of apoptotic cells are evident in Myc down-regulated cells exposed to gamma radiation for 72 hours compared with approximately 13% observed after only gamma radiation treatment. Consistent with the enhanced apoptosis is the inhibition of the MLH1 and MSH2 mismatch repair proteins, which, preventing the correction of ionizing radiation mismatches occurring during DNA replication, renders the cells more prone to radiation-induced apoptosis. CONCLUSIONS: Data herein reported show that Myc down-regulation lowers the apoptotic threshold in melanoma cells by inhibiting MLH1 and MSH2 proteins, thus increasing cell sensitivity to gamma radiation in a p53-independent fashion. Our results indicate the basis for developing new antitumoral therapeutic strategy, improving the management of melanoma patients.


Asunto(s)
Proliferación Celular/efectos de la radiación , Proteínas de Unión al ADN/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Apoptosis/efectos de la radiación , Disparidad de Par Base , Western Blotting , Proteínas Portadoras , Caspasa 3 , Caspasas/metabolismo , Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Análisis Mutacional de ADN , Reparación del ADN , Proteínas de Unión al ADN/genética , Relación Dosis-Respuesta en la Radiación , Regulación hacia Abajo , Citometría de Flujo , Rayos gamma , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/metabolismo , Melanoma/patología , Melanoma/radioterapia , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , ARN sin Sentido/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA