Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Bioinformatics ; 37(22): 4209-4215, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34048539

RESUMEN

MOTIVATION: Cardiomyocytes derived from stem cells are closely followed, notably since the discovery in 2007 of human induced pluripotent stem cells (hiPSC). Cardiomyocytes (hiPSC-CM) derived from hiPSC are indeed more and more used to study specific cardiac diseases as well as for developing novel applications such as drug safety experiments. Robust dedicated tools to characterize hiPSC-CM are now required. The hiPSC-CM morphology constitutes an important parameter since these cells do not demonstrate the expected rod shape, characteristic of native human cardiomyocytes. Similarly, the presence, the density and the organization of contractile structures would be a valuable parameter to study. Precise measurements of such characteristics would be useful in many situations: for describing pathological conditions, for pharmacological screens or even for studies focused on the hiPSC-CM maturation process. RESULTS: For this purpose, we developed a MATLAB based image analysis toolbox, which gives accurate values for cellular morphology parameters as well as for the contractile cell organization. AVAILABILITY AND IMPLEMENTATION: To demonstrate the power of this automated image analysis, we used a commercial maturation medium intended to promote the maturation status of hiPSC-CM, and compare the parameters with the ones obtained with standard culture medium, and with freshly dissociated mouse cardiomyocytes. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Humanos , Animales , Ratones , Células Cultivadas
3.
Clin Transl Med ; 11(3): e319, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33784018

RESUMEN

BACKGROUND: Severe ventricular rhythm disturbances are the hallmark of arrhythmogenic cardiomyopathy (ACM), and are often explained by structural conduction abnormalities. However, comprehensive investigations of ACM cell electrical instability are lacking. This study aimed to elucidate early electrical myogenic signature of ACM. METHODS: We investigated a 41-year-old ACM patient with a missense mutation (c.394C>T) in the DSC2 gene, which encodes desmocollin 2. Pathogenicity of this variant was confirmed using a zebrafish DSC2 model system. Control and DSC2 patient-derived pluripotent stem cells were reprogrammed and differentiated into cardiomyocytes (hiPSC-CM) to examine the specific electromechanical phenotype and its modulation by antiarrhythmic drugs (AADs). Samples of the patient's heart and hiPSC-CM were examined to identify molecular and cellular alterations. RESULTS: A shortened action potential duration was associated with reduced Ca2+ current density and increased K+ current density. This finding led to the elucidation of previously unknown abnormal repolarization dynamics in ACM patients. Moreover, the Ca2+ mobilised during transients was decreased, and the Ca2+ sparks frequency was increased. AAD testing revealed the following: (1) flecainide normalised Ca2+ transients and significantly decreased Ca2+ spark occurrence and (2) sotalol significantly lengthened the action potential and normalised the cells' contractile properties. CONCLUSIONS: Thorough analysis of hiPSC-CM derived from the DSC2 patient revealed abnormal repolarization dynamics, prompting the discovery of a short QT interval in some ACM patients. Overall, these results confirm a myogenic origin of ACM electrical instability and provide a rationale for prescribing class 1 and 3 AADs in ACM patients with increased ventricular repolarization reserve.


Asunto(s)
Antiarrítmicos/uso terapéutico , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/genética , Desmocolinas/genética , Electrocardiografía/métodos , Canales Iónicos/genética , Adulto , Animales , Arritmias Cardíacas/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Mutación Missense/genética , Pez Cebra
4.
Neuromuscul Disord ; 31(5): 419-430, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33752935

RESUMEN

Following the 2017 approval of a first spinal muscular atrophy (SMA) treatment by the European Medicines Agency, SMA Europe launched a Europe-wide survey with the goal of understanding patients' treatment expectations, realities of daily living and access to clinical trials and therapy, and how this varied according to parameters such as age and disease severity. A response rate of 31% yielded 1474 completed surveys from 26 European countries. In line with findings from a 2015 SMA Europe-led survey, participants considered stabilization of their condition to be progress. Notably, responses indicated that the current classification of SMA at diagnosis by 'type' often does not reflect current mobility level. Large gaps in treatment access were identified that varied in particular between age and disease severity groups, yet there was high interest in clinical trial participation. In addition, alternative treatment options, including combination therapies, are now expectations. These perspectives should be central considerations through the research and development processes of new SMA therapies, through data generation and discussions on access to therapies. Results from this survey indicate that collaboration between stakeholders is essential to the foundation upon which innovative approaches for SMA treatments and access can be explored.


Asunto(s)
Atrofia Muscular Espinal/psicología , Prioridad del Paciente , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Europa (Continente) , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Atrofia Muscular Espinal/terapia , Calidad de Vida , Índice de Severidad de la Enfermedad , Encuestas y Cuestionarios , Adulto Joven
5.
Orphanet J Rare Dis ; 14(1): 126, 2019 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-31174585

RESUMEN

BACKGROUND: Patient and public involvement for co-creation is increasingly recognized as a valuable strategy to develop healthcare research targeting patients' real needs. However, its practical implementation is not as advanced and unanimously accepted as it could be, due to cultural differences and complexities of managing healthcare programs and clinical studies, especially in the rare disease field. MAIN BODY: The European Neuromuscular Centre, a European foundation of patient organizations, involved its key stakeholders in a special workshop to investigate the position of the neuromuscular patient community with respect to healthcare and medical research to identify and address gaps and bottlenecks. The workshop took place in Milan (Italy) on January 19-20, 2018, involving 45 participants who were mainly representatives of the patient community, but also included experts from clinical centers, industry and regulatory bodies. In order to provide practical examples and constructive suggestions, specific topics were identified upfront. The first set of issues concerned the quality of life at specific phases of a patient's life, such as at the time of diagnosis or during pediatric to adult transition, and patient involvement in medical research on activities in daily living including patient reported outcome measures. The second set of issues concerned the involvement of patients in the management of clinical research tools, such as registries and biobanks, and their participation in study design or marketing authorization processes. Introductory presentations were followed by parallel working group sessions, to gain constructive contributions from all participants. The concept of shared decision making was used to ensure, in discussions, a partnership-based identification of the wishes and needs of all stakeholders involved, and the "ladder of participation" tool served as a model to evaluate the actual and the desired level of patients' involvement in all topics addressed. A general consensus on the outcome of the meeting was collected during the final plenary session. This paper reports the outcome of the workshop and the specific suggestions derived from the analysis of the first set of topics, related to quality of life. The outcomes of the second set of topics are reported elsewhere and are only briefly summarized herein for the sake of completeness. CONCLUSIONS: The neuromuscular community proved to be very active and engaged at different levels in the healthcare initiatives of interest. The workshop participants critically discussed several topics, providing practical examples where different stakeholders could play a role in making a change and bridging gaps. Overall, they indicated the need for education of all stakeholders for better communication, where everyone should become an ambassador to promote real change. Support should also come from institutions and healthcare bodies both at structural and economic level.


Asunto(s)
Toma de Decisiones , Enfermedades Neuromusculares/fisiopatología , Calidad de Vida , Investigación Biomédica , Humanos
6.
Sci Rep ; 8(1): 5618, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29618840

RESUMEN

LMNA gene encodes lamins A and C, two major components of the nuclear lamina, a network of intermediate filaments underlying the inner nuclear membrane. Most of LMNA mutations are associated with cardiac and/or skeletal muscles defects. Muscle laminopathies include Emery-Dreifuss Muscular Dystrophy, Limb-Girdle Muscular Dystrophy 1B, LMNA-related Congenital Muscular Dystrophy and Dilated Cardiomyopathy with conduction defects. To identify potential alterations in signaling pathways regulating muscle differentiation in LMNA-mutated myoblasts, we used a previously described model of conditionally immortalized murine myoblasts: H-2K cell lines. Comparing gene expression profiles in wild-type and Lmna∆8-11 H-2K myoblasts, we identified two major alterations in the BMP (Bone Morphogenetic Protein) pathway: Bmp4 downregulation and Smad6 overexpression. We demonstrated that these impairments lead to Lmna∆8-11 myoblasts premature differentiation and can be rescued by downregulating Smad6 expression. Finally, we showed that BMP4 pathway defects are also present in myoblasts from human patients carrying different heterozygous LMNA mutations.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular , Lamina Tipo A/metabolismo , Distrofia Muscular de Emery-Dreifuss/patología , Mutación , Mioblastos/patología , Proteína smad6/metabolismo , Adulto , Animales , Proteína Morfogenética Ósea 4/genética , Estudios de Casos y Controles , Humanos , Lamina Tipo A/genética , Masculino , Ratones , Ratones Noqueados , Desarrollo de Músculos , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/metabolismo , Mioblastos/metabolismo , Proteína smad6/genética
7.
Orphanet J Rare Dis ; 12(1): 147, 2017 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-28854936

RESUMEN

Since the identification of the first disease causing mutation in the gene coding for emerin, a transmembrane protein of the inner nuclear membrane, hundreds of mutations and variants have been found in genes encoding for nuclear envelope components. These proteins can be part of the inner nuclear membrane (INM), such as emerin or SUN proteins, outer nuclear membrane (ONM), such as Nesprins, or the nuclear lamina, such as lamins A and C. However, they physically interact with each other to insure the nuclear envelope integrity and mediate the interactions of the nuclear envelope with both the genome, on the inner side, and the cytoskeleton, on the outer side. The core of this complex, called LINC (LInker of Nucleoskeleton to Cytoskeleton) is composed of KASH and SUN homology domain proteins. SUN proteins are INM proteins which interact with lamins by their N-terminal domain and with the KASH domain of nesprins located in the ONM by their C-terminal domain.Although most of these proteins are ubiquitously expressed, their mutations have been associated with a large number of clinically unrelated pathologies affecting specific tissues. Moreover, variants in SUN proteins have been found to modulate the severity of diseases induced by mutations in other LINC components or interactors. For these reasons, the diagnosis and the identification of the molecular explanation of "nuclear envelopathies" is currently challenging.The aim of this review is to summarize the human diseases caused by mutations in genes coding for INM proteins, nuclear lamina, and ONM proteins, and to discuss their potential physiopathological mechanisms that could explain the large spectrum of observed symptoms.


Asunto(s)
Membrana Nuclear/metabolismo , Animales , Citoesqueleto/metabolismo , Humanos , Laminas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/metabolismo
8.
Mol Cell Biol ; 35(24): 4110-20, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26416879

RESUMEN

Acetylcholine receptor (AChR) expression in innervated muscle is limited to the synaptic region. Neuron-induced electrical activity participates in this compartmentalization by promoting the repression of AChR expression in the extrasynaptic regions. Here, we show that the corepressor CtBP1 (C-terminal binding protein 1) is present on the myogenin promoter together with repressive histone marks. shRNA-mediated downregulation of CtBP1 expression is sufficient to derepress myogenin and AChR expression in innervated muscle. Upon denervation, CtBP1 is displaced from the myogenin promoter and relocates to the cytoplasm, while repressive histone marks are replaced by activating ones concomitantly to the activation of myogenin expression. We also observed that upon denervation the p21-activated kinase 1 (PAK1) expression is upregulated, suggesting that phosphorylation by PAK1 may be involved in the relocation of CtBP1. Indeed, preventing CtBP1 Ser158 phosphorylation induces CtBP1 accumulation in the nuclei and abrogates the activation of myogenin and AChR expression. Altogether, these findings reveal a molecular mechanism to account for the coordinated control of chromatin modifications and muscle gene expression by presynaptic neurons via a PAK1/CtBP1 pathway.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Proteínas de Unión al ADN/metabolismo , Desnervación Muscular , Músculo Esquelético/inervación , Miogenina/biosíntesis , Quinasas p21 Activadas/metabolismo , Transporte Activo de Núcleo Celular/genética , Oxidorreductasas de Alcohol/genética , Animales , Núcleo Celular/metabolismo , Cromatina/genética , Cromatina/metabolismo , Citoplasma/metabolismo , Proteínas de Unión al ADN/genética , Histonas/genética , Histonas/metabolismo , Ratones , Músculo Esquelético/metabolismo , Miogenina/genética , Fosforilación , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Interferente Pequeño , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Activación Transcripcional/genética , Regulación hacia Arriba , Quinasas p21 Activadas/biosíntesis , Quinasas p21 Activadas/genética
9.
J Biol Chem ; 290(7): 4215-24, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25516595

RESUMEN

Skeletal muscle atrophy is a severe condition of muscle mass loss. Muscle atrophy is caused by a down-regulation of protein synthesis and by an increase of protein breakdown due to the ubiquitin-proteasome system and autophagy activation. Up-regulation of specific genes, such as the muscle-specific E3 ubiquitin ligase MAFbx, by FoxO transcription factors is essential to initiate muscle protein ubiquitination and degradation during atrophy. HDAC6 is a particular HDAC, which is functionally related to the ubiquitin proteasome system via its ubiquitin binding domain. We show that HDAC6 is up-regulated during muscle atrophy. HDAC6 activation is dependent on the transcription factor FoxO3a, and the inactivation of HDAC6 in mice protects against muscle wasting. HDAC6 is able to interact with MAFbx, a key ubiquitin ligase involved in muscle atrophy. Our findings demonstrate the implication of HDAC6 in skeletal muscle wasting and identify HDAC6 as a new downstream target of FoxO3a in stress response. This work provides new insights in skeletal muscle atrophy development and opens interesting perspectives on HDAC6 as a valuable marker of muscle atrophy and a potential target for pharmacological treatments.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/patología , Animales , Western Blotting , Células Cultivadas , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Histona Desacetilasa 6 , Histona Desacetilasas/química , Histona Desacetilasas/genética , Humanos , Inmunoprecipitación , Integrasas/metabolismo , Ratones , Ratones Noqueados , Desnervación Muscular , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
J Vis Exp ; (79): e50610, 2013 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-24084155

RESUMEN

The Drosophila eye is widely used as a model for studies of development and neuronal degeneration. With the powerful mitotic recombination technique, elegant genetic screens based on clonal analysis have led to the identification of signaling pathways involved in eye development and photoreceptor (PR) differentiation at larval stages. We describe here the Tomato/GFP-FLP/FRT method, which can be used for rapid clonal analysis in the eye of living adult Drosophila. Fluorescent photoreceptor cells are imaged with the cornea neutralization technique, on retinas with mosaic clones generated by flipase-mediated recombination. This method has several major advantages over classical histological sectioning of the retina: it can be used for high-throughput screening and has proved an effective method for identifying the factors regulating PR survival and function. It can be used for kinetic analyses of PR degeneration in the same living animal over several weeks, to demonstrate the requirement for specific genes for PR survival or function in the adult fly. This method is also useful for addressing cell autonomy issues in developmental mutants, such as those in which the establishment of planar cell polarity is affected.


Asunto(s)
Drosophila/citología , Proteínas Fluorescentes Verdes/química , Células Fotorreceptoras de Invertebrados/citología , Retina/citología , Animales , Drosophila/genética , Proteínas de Drosophila , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Microscopía Fluorescente/métodos , Células Fotorreceptoras de Invertebrados/química , Células Fotorreceptoras de Invertebrados/metabolismo , Recombinación Genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/química , Factores de Transcripción/genética
11.
Nucleus ; 1(1): 40-52, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21327104

RESUMEN

The cell nucleus communicates with the rest of the cell via nucleo/cytoplasmic transport of proteins and RNA through the nuclear pores. Direct mechanical links between the nucleus and the cytoplasm have recently emerged in the form of LINC (Linkers of the nucleoskeleton to the cytoskeleton) protein complexes. A LINC complex consists of four components. At its core are an inner nuclear membrane (INM) transmembrane protein and an outer nuclear membrane (ONM) transmembrane protein which physically interact with each other in the lumen of the NE. The INM LINC component interacts on the nucleoplasmic side with either the lamina or with an INM-associated protein. The ONM LINC component on the other hand contacts on the cytoplasmatic side a component of the cytoskeleton. This review highlights the components of LINC complexes and their emerging roles in mechanotransduction, nuclear migration, chromosome positioning, signaling, meiosis, cytoskeletal organization and human disease.


Asunto(s)
Proteínas de la Membrana/metabolismo , Proteínas Nucleares/metabolismo , Animales , Núcleo Celular/metabolismo , Citoesqueleto/metabolismo , Humanos , Laminas/genética , Laminas/metabolismo , Distrofia Muscular de Emery-Dreifuss/metabolismo , Distrofia Muscular de Emery-Dreifuss/patología , Membrana Nuclear/metabolismo
12.
J Cell Biol ; 184(1): 31-44, 2009 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-19124654

RESUMEN

The LMNA gene encodes lamins A and C, two intermediate filament-type proteins that are important determinants of interphase nuclear architecture. Mutations in LMNA lead to a wide spectrum of human diseases including autosomal dominant Emery-Dreifuss muscular dystrophy (AD-EDMD), which affects skeletal and cardiac muscle. The cellular mechanisms by which mutations in LMNA cause disease have been elusive. Here, we demonstrate that defects in neuromuscular junctions (NMJs) are part of the disease mechanism in AD-EDMD. Two AD-EDMD mouse models show innervation defects including misexpression of electrical activity-dependent genes and altered epigenetic chromatin modifications. Synaptic nuclei are not properly recruited to the NMJ because of mislocalization of nuclear envelope components. AD-EDMD patients with LMNA mutations show the same cellular defects as the AD-EDMD mouse models. These results suggest that lamin A/C-mediated NMJ defects contribute to the AD-EDMD disease phenotype and provide insights into the cellular and molecular mechanisms for the muscle-specific phenotype of AD-EDMD.


Asunto(s)
Lamina Tipo A/fisiología , Distrofia Muscular de Emery-Dreifuss/metabolismo , Unión Neuromuscular/patología , Animales , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Modelos Animales de Enfermedad , Humanos , Lamina Tipo A/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Desnervación Muscular , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/ultraestructura , Músculo Esquelético/inervación , Músculo Esquelético/ultraestructura , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/patología , Transporte de Proteínas
13.
Am J Physiol Cell Physiol ; 289(4): C908-17, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15930144

RESUMEN

We examined whether calcineurin-NFAT (nuclear factors of activated T cells) signaling plays a role in specifically directing the expression of utrophin in the synaptic compartment of muscle fibers. Immunofluorescence experiments revealed the accumulation of components of the calcineurin-NFAT signaling cascade within the postsynaptic membrane domain of the neuromuscular junction. RT-PCR analysis using synaptic vs. extrasynaptic regions of muscle fibers confirmed these findings by showing an accumulation of calcineurin transcripts within the synaptic compartment. We also examined the effect of calcineurin on utrophin gene expression. Pharmacological inhibition of calcineurin in mice with either cyclosporin A or FK506 resulted in a marked decrease in utrophin A expression at synaptic sites, whereas constitutive activation of calcineurin had the opposite effect. Mutation of the previously identified NFAT binding site in the utrophin A promoter region, followed by direct gene transfer studies in mouse muscle, led to an inhibition in the synaptic expression of a lacZ reporter gene construct. Transfection assays performed with cultured myogenic cells indicated that calcineurin acted additively with GA binding protein (GABP) to transactivate utrophin A gene expression. Because both GABP- and calcineurin-mediated pathways are targeted by peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha), we examined whether this coactivator contributes to utrophin gene expression. In vitro and in vivo transfection experiments showed that PGC-1alpha alone induces transcription from the utrophin A promoter. Interestingly, this induction is largely potentiated by coexpression of PGC-1alpha with GABP. Together, these studies indicate that the synaptic expression of utrophin is also driven by calcineurin-NFAT signaling and occurs in conjunction with signaling events that involve GABP and PGC-1alpha.


Asunto(s)
Calcineurina/fisiología , Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica/fisiología , Unión Neuromuscular/metabolismo , Proteínas Nucleares/fisiología , Transactivadores/fisiología , Factores de Transcripción/fisiología , Utrofina/biosíntesis , Animales , Ciclosporina/farmacología , Factor de Transcripción de la Proteína de Unión a GA , Inmunosupresores/farmacología , Ratones , Músculo Esquelético/metabolismo , Factores de Transcripción NFATC , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Transducción de Señal , Tacrolimus/farmacología
14.
Nat Neurosci ; 8(3): 313-21, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15711539

RESUMEN

Electrical activity arising from motor innervation influences skeletal muscle physiology by controlling the expression of many muscle genes, including those encoding acetylcholine receptor (AChR) subunits. How electrical activity is converted into a transcriptional response remains largely unknown. We show that motor innervation controls chromatin acetylation in skeletal muscle and that histone deacetylase 9 (HDAC9) is a signal-responsive transcriptional repressor which is downregulated upon denervation, with consequent upregulation of chromatin acetylation and AChR expression. Forced expression of Hdac9 in denervated muscle prevents upregulation of activity-dependent genes and chromatin acetylation by linking myocyte enhancer factor 2 (MEF2) and class I HDACs. By contrast, Hdac9-null mice are supersensitive to denervation-induced changes in gene expression and show chromatin hyperacetylation and delayed perinatal downregulation of myogenin, an activator of AChR genes. These findings show a molecular mechanism to account for the control of chromatin acetylation by presynaptic neurons and the activity-dependent regulation of skeletal muscle genes by motor innervation.


Asunto(s)
Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Expresión Génica/fisiología , Histona Desacetilasas/metabolismo , Músculo Esquelético/fisiología , Neuronas/fisiología , Proteínas Represoras/metabolismo , Acetilación , Factores de Edad , Animales , Animales Recién Nacidos , Western Blotting/métodos , Clonación Molecular , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Electroporación/métodos , Embrión de Mamíferos , Técnica del Anticuerpo Fluorescente/métodos , Proteínas Fluorescentes Verdes/metabolismo , Histona Desacetilasas/clasificación , Histona Desacetilasas/deficiencia , Histona Desacetilasas/genética , Histonas/metabolismo , Inmunoprecipitación/métodos , Factores de Transcripción MEF2 , Ratones , Ratones Noqueados , Desnervación Muscular/métodos , Músculo Esquelético/inervación , Factores Reguladores Miogénicos , Miogenina/metabolismo , ARN Mensajero/biosíntesis , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factores de Tiempo , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
15.
Proc Natl Acad Sci U S A ; 101(52): 18189-94, 2004 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-15604144

RESUMEN

The muscle-specific receptor tyrosine kinase (MuSK) is part of a receptor complex, activated by neural agrin, that orchestrates the differentiation of the neuromuscular junction (NMJ). To gain insight into the function of the MuSK complex, we have developed a proteomic approach to identify new MuSK partners. MS analysis of MuSK crosslink products from postsynaptic membranes of the Torpedo electrocytes identified the adaptor protein 14-3-3 gamma. The 14-3-3 gamma protein was found localized at the adult rat NMJ. Cotransfection experiments in COS-7 cells showed that MuSK codistributed with the 14-3-3 gamma protein at the plasma membrane. Furthermore, 14-3-3 gamma was copurified by affinity chromatography with MuSK from transfected COS-7 cells and myotubes. The 14-3-3 gamma protein did not colocalize with agrin-elicited acetylcholine receptor (AChR) aggregates in cultured myotubes, suggesting that it is not involved in AChR clustering. Expression of 14-3-3 gamma specifically repressed the transcription of several synaptic reporter genes in cultured myotubes. This repression was potentiated by MuSK expression. Moreover, the expression of 14-3-3 gamma in muscle fibers in vivo caused both the repression of synaptic genes transcription and morphological perturbations of the NMJ. Our data extend the notion that, apart from its well documented role in AChR clustering, the MuSK complex might also be involved in the regulation of synaptic gene expression at the NMJ.


Asunto(s)
Proteínas 14-3-3/fisiología , Unión Neuromuscular/fisiología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Colinérgicos/metabolismo , Sinapsis/patología , Transcripción Genética , Proteínas 14-3-3/metabolismo , Agrina/metabolismo , Animales , Células COS , Membrana Celular/metabolismo , Células Cultivadas , Cromatografía , Reactivos de Enlaces Cruzados , Electrólitos/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Vectores Genéticos , Proteínas Fluorescentes Verdes/metabolismo , Inmunoprecipitación , Microscopía Fluorescente , Músculos/metabolismo , Neuronas/metabolismo , Unión Proteica , Proteómica , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Factores de Tiempo , Torpedo , Transfección
16.
Ann N Y Acad Sci ; 998: 53-65, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-14592863

RESUMEN

Agrin is the key neural factor that controls muscle postsynaptic differentiation, including the induction of synapse-specific transcription via neuregulins. In 1995, the promoter element responsible for the targeting of AChR delta and epsilon gene transcription to the skeletal muscle subsynaptic area was identified. This element, named N-box, recruits the Ets-related transcription factor GABP to AChR delta and epsilon promoters, and both the N-box and GABP are required to obtain transcriptional stimulation by neuregulins. The physiological importance of the N-box has been definitively established with the discovery of myasthenic families carrying single-point mutations in the N-box of the AChR epsilon gene promoter and showing reduced levels of AChR epsilon subunit expression. The control of synapse-specific transcription by agrin and neuregulins through the N-box and GABP is not restricted to the case of AChR genes. The same regulation holds true for the ACh esterase and utrophin genes, thus showing that nerve-induced transcriptional activation of several synapse-specific genes is triggered by a common mechanism involving agrin, neuregulins, and ultimately the N-box and Ets-related transcription factors.


Asunto(s)
Expresión Génica , Unión Neuromuscular/genética , Sinapsis/genética , Agrina/metabolismo , Animales , Proteínas de Unión al ADN/genética , Humanos , Proteínas de la Membrana , Neurregulinas/metabolismo , Regiones Promotoras Genéticas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Colinérgicos/metabolismo , Transducción de Señal , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...