Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Development ; 148(2)2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33298462

RESUMEN

Formation of skeletal muscle is among the most striking examples of cellular plasticity in animal tissue development, and while muscle progenitor cells are reprogrammed by epithelial-mesenchymal transition (EMT) to migrate during embryonic development, the regulation of EMT in post-natal myogenesis remains poorly understood. Here, we demonstrate that the long noncoding RNA (lncRNA) Meg3 regulates EMT in myoblast differentiation and skeletal muscle regeneration. Chronic inhibition of Meg3 in C2C12 myoblasts induced EMT, and suppressed cell state transitions required for differentiation. Furthermore, adenoviral Meg3 knockdown compromised muscle regeneration, which was accompanied by abnormal mesenchymal gene expression and interstitial cell proliferation. Transcriptomic and pathway analyses of Meg3-depleted C2C12 myoblasts and injured skeletal muscle revealed a significant dysregulation of EMT-related genes, and identified TGFß as a key upstream regulator. Importantly, inhibition of TGFßR1 and its downstream effectors, and the EMT transcription factor Snai2, restored many aspects of myogenic differentiation in Meg3-depleted myoblasts in vitro We further demonstrate that reduction of Meg3-dependent Ezh2 activity results in epigenetic alterations associated with TGFß activation. Thus, Meg3 regulates myoblast identity to facilitate progression into differentiation.


Asunto(s)
Plasticidad de la Célula/genética , Transición Epitelial-Mesenquimal/genética , Mioblastos/citología , Mioblastos/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Adhesión Celular/genética , Diferenciación Celular/genética , Línea Celular , Movimiento Celular/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Técnicas de Silenciamiento del Gen , Histonas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Mesodermo/patología , Metilación , Ratones , Mitocondrias/metabolismo , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Mutación/genética , ARN Largo no Codificante/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Regeneración , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
J Biol Chem ; 292(25): 10613-10629, 2017 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-28473466

RESUMEN

Cardiomyocytes acquire their primary specialized function (contraction) before exiting the cell cycle. In this regard, proliferation and differentiation must be precisely coordinated for proper cardiac morphogenesis. Here, we have investigated the complex transcriptional mechanisms employed by cardiomyocytes to coordinate antagonistic cell-cycle and differentiation gene programs through the molecular dissection of the core cardiac transcription factor, MEF2. Knockdown of individual MEF2 proteins, MEF2A, -C, and -D, in primary neonatal cardiomyocytes resulted in radically distinct and opposite effects on cellular homeostasis and gene regulation. MEF2A and MEF2D were absolutely required for cardiomyocyte survival, whereas MEF2C, despite its major role in cardiac morphogenesis and direct reprogramming, was dispensable for this process. Inhibition of MEF2A or -D also resulted in the activation of cell-cycle genes and down-regulation of markers of terminal differentiation. In striking contrast, the regulation of cell-cycle and differentiation gene programs by MEF2C was antagonistic to that of MEF2A and -D. Computational analysis of regulatory regions from MEF2 isoform-dependent gene sets identified the Notch and Hedgehog signaling pathways as key determinants in coordinating MEF2 isoform-specific control of antagonistic gene programs. These results reveal that mammalian MEF2 family members have distinct transcriptional functions in cardiomyocytes and suggest that these differences are critical for proper development and maturation of the heart. Analysis of MEF2 isoform-specific function in neonatal cardiomyocytes has yielded insight into an unexpected transcriptional regulatory mechanism by which these specialized cells utilize homologous members of a core cardiac transcription factor to coordinate cell-cycle and differentiation gene programs.


Asunto(s)
Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Miocitos Cardíacos/citología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley
3.
J Biol Chem ; 292(51): 20975-20988, 2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29054930

RESUMEN

The distinct morphological and functional properties of the cardiac chambers arise from an elaborate developmental program involving cell lineage determination, morphogenesis, and dynamic spatiotemporal gene expression patterns. Although a number of transcription factors have been identified for proper gene regulation in the chambers, the complete transcriptional network that controls these patterns remains poorly defined. Previous studies have implicated the MEF2C transcription factor in the regulation of chamber-restricted enhancers. To better understand the mechanisms of MEF2-mediated regional gene regulation in the heart, we took advantage of MEF2A knock-out (KO) mice, a model that displays a predominantly ventricular chamber phenotype. Transcriptomic analysis of atrial and ventricular tissue from adult MEF2A KO hearts revealed a striking difference in chamber gene expression, with a larger proportion of dysregulated genes in the atrial chambers. Canonical pathway analysis of genes preferentially dysregulated in the atria and ventricles revealed distinct MEF2A-dependent cellular processes in each cardiac chamber. In the atria, MEF2A regulated genes involved in fibrosis and adhesion, whereas in the ventricles, it controlled inflammation and endocytosis. Finally, analysis of transcription factor-binding site motifs of differentially dysregulated genes uncovered distinct MEF2A co-regulators for the atrial and ventricular gene sets, and a subset of these was found to cooperate with MEF2A. In conclusion, our results suggest a mechanism in which MEF2 transcriptional activity is differentially recruited to fine-tune gene expression levels in each cardiac chamber. This regulatory mechanism ensures optimal output of these gene products for proper physiological function of the atrial and ventricular chambers.


Asunto(s)
Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/metabolismo , Animales , Regulación de la Expresión Génica , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Factores de Transcripción MEF2/deficiencia , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Tisular
4.
J Biol Chem ; 290(38): 23162-72, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26240138

RESUMEN

Understanding cell cycle regulation in postmitotic cardiomyocytes may lead to new therapeutic approaches to regenerate damaged cardiac tissue. We have demonstrated previously that microRNAs encoded by the Gtl2-Dio3 noncoding RNA locus function downstream of the MEF2A transcription factor in skeletal muscle regeneration. We have also reported expression of these miRNAs in the heart. Here we investigated the role of two Gtl2-Dio3 miRNAs, miR-410 and miR-495, in cardiac muscle. Overexpression of miR-410 and miR-495 robustly stimulated cardiomyocyte DNA synthesis and proliferation. Interestingly, unlike our findings in skeletal muscle, these miRNAs did not modulate the activity of the WNT signaling pathway. Instead, these miRNAs targeted Cited2, a coactivator required for proper cardiac development. Consistent with miR-410 and miR-495 overexpression, siRNA knockdown of Cited2 in neonatal cardiomyocytes resulted in robust proliferation. This phenotype was associated with reduced expression of Cdkn1c/p57/Kip2, a cell cycle inhibitor, and increased expression of VEGFA, a growth factor with proliferation-promoting effects. Therefore, miR-410 and miR-495 are among a growing number of miRNAs that have the ability to potently stimulate neonatal cardiomyocyte proliferation.


Asunto(s)
Proliferación Celular/fisiología , Sitios Genéticos/fisiología , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Ratones , MicroARNs/genética , Miocardio/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/genética , Transactivadores/genética , Factores de Transcripción/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Vía de Señalización Wnt/fisiología
5.
J Biol Chem ; 290(40): 24367-80, 2015 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-26294766

RESUMEN

The cardiomyocyte cell cycle is a poorly understood process. Mammalian cardiomyocytes permanently withdraw from the cell cycle shortly after birth but can re-enter the cell cycle and proliferate when subjected to injury within a brief temporal window in the neonatal period. Thus, investigating the mechanisms of cell cycle regulation in neonatal cardiomyocytes may provide critical insight into the molecular events that prevent adult myocytes from proliferating in response to injury or stress. MEF2D is a key transcriptional mediator of pathological remodeling in the adult heart downstream of various stress-promoting insults. However, the specific gene programs regulated by MEF2D in cardiomyocytes are unknown. By performing genome-wide transcriptome analysis using MEF2D-depleted neonatal cardiomyocytes, we found a significant impairment in the cell cycle, characterized by the up-regulation of numerous positive cell cycle regulators. Expression of Pten, the primary negative regulator of PI3K/Akt, was significantly reduced in MEF2D-deficient cardiomyocytes and found to be a direct target gene of MEF2D. Consistent with these findings mutant cardiomyocytes showed activation of the PI3K/Akt survival pathway. Paradoxically, prolonged deficiency of MEF2D in neonatal cardiomyocytes did not trigger proliferation but instead resulted in programmed cell death, which is likely mediated by the E2F transcription factor. These results demonstrate a critical role for MEF2D in cell cycle regulation of post-mitotic, neonatal cardiomyocytes in vitro.


Asunto(s)
Miocitos Cardíacos/citología , Animales , Animales Recién Nacidos , Apoptosis , Caspasa 3/metabolismo , Ciclo Celular , Proliferación Celular , Supervivencia Celular , Factores de Transcripción E2F/metabolismo , Fibroblastos/metabolismo , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/fisiología , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Transcriptoma
6.
J Biol Chem ; 290(2): 1256-68, 2015 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-25416778

RESUMEN

Skeletal muscle differentiation requires precisely coordinated transcriptional regulation of diverse gene programs that ultimately give rise to the specialized properties of this cell type. In Drosophila, this process is controlled, in part, by MEF2, the sole member of an evolutionarily conserved transcription factor family. By contrast, vertebrate MEF2 is encoded by four distinct genes, Mef2a, -b, -c, and -d, making it far more challenging to link this transcription factor to the regulation of specific muscle gene programs. Here, we have taken the first step in molecularly dissecting vertebrate MEF2 transcriptional function in skeletal muscle differentiation by depleting individual MEF2 proteins in myoblasts. Whereas MEF2A is absolutely required for proper myoblast differentiation, MEF2B, -C, and -D were found to be dispensable for this process. Furthermore, despite the extensive redundancy, we show that mammalian MEF2 proteins regulate a significant subset of nonoverlapping gene programs. These results suggest that individual MEF2 family members are able to recognize specific targets among the entire cohort of MEF2-regulated genes in the muscle genome. These findings provide opportunities to modulate the activity of MEF2 isoforms and their respective gene programs in skeletal muscle homeostasis and disease.


Asunto(s)
Diferenciación Celular/genética , Evolución Molecular , Factores de Transcripción MEF2/biosíntesis , Músculo Esquelético/crecimiento & desarrollo , Isoformas de Proteínas/biosíntesis , Animales , Células COS , Chlorocebus aethiops , Drosophila/genética , Drosophila/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción MEF2/antagonistas & inhibidores , Factores de Transcripción MEF2/genética , Mamíferos/genética , Mamíferos/crecimiento & desarrollo , Ratones , Desarrollo de Músculos/genética , Mioblastos/citología , Mioblastos/metabolismo , Isoformas de Proteínas/genética
7.
Development ; 140(1): 31-42, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23154418

RESUMEN

Understanding the molecular mechanisms of skeletal muscle regeneration is crucial to exploiting this pathway for use in tissue repair. Our data demonstrate that the MEF2A transcription factor plays an essential role in skeletal muscle regeneration in adult mice. Injured Mef2a knockout mice display widespread necrosis and impaired myofiber formation. MEF2A controls this process through its direct regulation of the largest known mammalian microRNA (miRNA) cluster, the Gtl2-Dio3 locus. A subset of the Gtl2-Dio3 miRNAs represses secreted Frizzled-related proteins (sFRPs), inhibitors of WNT signaling. Consistent with these data, Gtl2-Dio3-encoded miRNAs are downregulated in regenerating Mef2a knockout muscle, resulting in upregulated sFRP expression and attenuated WNT activity. Furthermore, myogenic differentiation in Mef2a-deficient myoblasts is rescued by overexpression of miR-410 and miR-433, two miRNAs in the Gtl2-Dio3 locus that repress sFRP2, or by treatment with recombinant WNT3A and WNT5A. Thus, miRNA-mediated modulation of WNT signaling by MEF2A is a requisite step for proper muscle regeneration, and represents an attractive pathway for enhancing regeneration of diseased muscle.


Asunto(s)
Carbocianinas/metabolismo , MicroARNs/metabolismo , Músculo Esquelético/fisiología , Factores Reguladores Miogénicos/fisiología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Regeneración/fisiología , Proteínas Wnt/metabolismo , Animales , Células COS , Línea Celular , Células Cultivadas , Chlorocebus aethiops , Receptores Frizzled/genética , Técnicas de Silenciamiento del Gen , Humanos , Factores de Transcripción MEF2 , Ratones , Ratones Noqueados , Factores Reguladores Miogénicos/genética , Transducción de Señal/fisiología , Regulación hacia Arriba/genética , Proteínas Wnt/fisiología
8.
Cell Mol Life Sci ; 71(9): 1641-56, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24218011

RESUMEN

Structural abnormalities in striated muscle have been observed in numerous transcription factor gain- and loss-of-function phenotypes in animal and cell culture model systems, indicating that transcription is important in regulating the cytoarchitecture. While most characterized cytoarchitectural defects are largely indistinguishable by histological and ultrastructural criteria, analysis of dysregulated gene expression in each mutant phenotype has yielded valuable information regarding specific structural gene programs that may be uniquely controlled by each of these transcription factors. Linking the formation and maintenance of each subcellular structure or subset of proteins within a cytoskeletal compartment to an overlapping but distinct transcription factor cohort may enable striated muscle to control cytoarchitectural function in an efficient and specific manner. Here we summarize the available evidence that connects transcription factors, those with established roles in striated muscle such as MEF2 and SRF, as well as other non-muscle transcription factors, to the regulation of a defined cytoskeletal structure. The notion that genes encoding proteins localized to the same subcellular compartment are coordinately transcriptionally regulated may prompt rationally designed approaches that target specific transcription factor pathways to correct structural defects in muscle disease.


Asunto(s)
Costameras/metabolismo , Redes Reguladoras de Genes , Sarcómeros/metabolismo , Animales , Costameras/genética , Citoesqueleto/química , Citoesqueleto/metabolismo , Humanos , Músculo Esquelético/metabolismo , Miocitos Cardíacos/metabolismo , Sarcómeros/genética , Factores de Transcripción/metabolismo
9.
Proc Natl Acad Sci U S A ; 107(42): 18097-102, 2010 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-20923879

RESUMEN

Unknown molecular responses to sarcomere protein gene mutations account for pathologic remodeling in hypertrophic cardiomyopathy (HCM), producing myocyte growth and increased cardiac fibrosis. To determine if hypertrophic signals activated myocyte enhancer factor-2 (Mef2), we studied mice carrying the HCM mutation, myosin heavy-chain Arg403Gln, (MHC(403/+)) and an Mef2-dependent ß-galactosidase reporter transgene. In young, prehypertrophic MHC(403/+) mice the reporter was not activated. In hypertrophic hearts, activation of the Mef2-dependent reporter was remarkably heterogeneous and was observed consistently in myocytes that bordered fibrotic foci with necrotic cells, MHC(403/+) myocytes with Mef2-dependent reporter activation reexpressed the fetal myosin isoform (ßMHC), a molecular marker of hypertrophy, although MHC(403/+) myocytes with or without ßMHC expression were comparably enlarged over WT myocytes. To consider Mef2 roles in severe HCM, we studied homozygous MHC(403/403) mice, which have accelerated remodeling, widespread myocyte necrosis, and neonatal lethality. Levels of phosphorylated class II histone deacetylases that activate Mef2 were substantially increased in MHC(403/403) hearts, but Mef2-dependent reporter activation was patchy. Sequential analyses showed myocytes increased Mef2-dependent reporter activity before death. Our data dissociate myocyte hypertrophy, a consistent response in HCM, from heterogeneous Mef2 activation and reexpression of a fetal gene program. The temporal and spatial relationship of Mef2-dependent gene activation with myocyte necrosis and fibrosis in MHC(403/+) and MHC(403/403) hearts defines Mef2 activation as a molecular signature of stressed HCM myocytes that are poised to die.


Asunto(s)
Cardiomiopatía Hipertrófica/patología , Factores Reguladores Miogénicos/metabolismo , Animales , Western Blotting , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Fibrosis , Genes Reporteros , Factores de Transcripción MEF2 , Ratones , Factores Reguladores Miogénicos/genética , Necrosis , Fosforilación , Mutación Puntual
10.
J Biol Chem ; 286(34): 29644-53, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21724844

RESUMEN

The Mef2 family of transcription factors regulates muscle differentiation, but the specific gene programs controlled by each member remain unknown. Characterization of Mef2A knock-out mice has revealed severe myofibrillar defects in cardiac muscle indicating a requirement for Mef2A in cytoarchitectural integrity. Through comprehensive expression analysis of Mef2A-deficient hearts, we identified a cohort of dysregulated genes whose products localize to the peripheral Z-disc/costamere region. Many of these genes are essential for costamere integrity and function. Here we demonstrate that these genes are directly regulated by Mef2A, establishing a mechanism by which Mef2A controls the costamere. In an independent model system, acute knockdown of Mef2A in primary neonatal cardiomyocytes resulted in profound malformations of myofibrils and focal adhesions accompanied by adhesion-dependent programmed cell death. These findings indicate a role for Mef2A in cardiomyocyte survival through regulation of costamere integrity. Finally, bioinformatics analysis identified over-represented transcription factor-binding sites in this network of costamere promoters that may provide insight into the mechanism by which costamere genes are regulated by Mef2A. The global control of costamere gene expression adds another dimension by which this essential macromolecular complex may be regulated in health and disease.


Asunto(s)
Costameras/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas de Dominio MADS/metabolismo , Miocardio/metabolismo , Factores Reguladores Miogénicos/metabolismo , Elementos de Respuesta/fisiología , Animales , Células COS , Chlorocebus aethiops , Costameras/genética , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , Proteínas de Dominio MADS/genética , Factores de Transcripción MEF2 , Ratones , Ratones Noqueados , Factores Reguladores Miogénicos/genética , Ratas , Ratas Sprague-Dawley
11.
FASEB J ; 25(7): 2276-86, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21427212

RESUMEN

The calcium-calmodulin-regulated protein phosphatase calcineurin plays an important regulatory role in muscle differentiation, fiber-type determination, hypertrophy, and muscle regeneration. Because calcineurin functions in numerous processes in muscle, multiple mechanisms are likely necessary to ensure that the activity of this phosphatase is appropriately regulated. Here we demonstrate that the muscle-specific scaffolding protein myospryn modulates calcineurin signaling by inhibiting calcineurin-dependent transcriptional activity in C2C12 myoblasts through direct interaction with the enzyme via its noncanonical tripartite motif (TRIM-like). Consistent with these data, transgenic mice overexpressing both the TRIM-like domain of myospryn and constitutively active calcineurin displayed a severe attenuation in the ability of calcineurin to induce a slow-fiber phenotype. Furthermore, transgenic mice overexpressing the TRIM-like domain of myospryn displayed attenuated muscle regeneration after cardiotoxin-induced muscle injury. These results indicate that myospryn functions as a novel inhibitor of the calcineurin signaling pathway in skeletal muscle.


Asunto(s)
Calcineurina/metabolismo , Proteínas Portadoras/metabolismo , Fibras Musculares de Contracción Lenta/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Animales , Animales Recién Nacidos , Sitios de Unión , Células COS , Calcineurina/genética , Proteínas Portadoras/genética , Línea Celular , Células Cultivadas , Chlorocebus aethiops , Proteínas Cardiotóxicas de Elápidos/toxicidad , Crotoxina/toxicidad , Combinación de Medicamentos , Immunoblotting , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Transgénicos , Proteínas Musculares/genética , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Unión Proteica , Ratas , Regeneración
12.
Circ Res ; 106(5): 952-60, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20093629

RESUMEN

RATIONALE: The vasoactive peptide angiotensin II (Ang II) is a potent cardiotoxic hormone whose actions have been well studied, yet questions remain pertaining to the downstream factors that mediate its effects in cardiomyocytes. OBJECTIVE: The in vivo role of the myocyte enhancer factor (MEF)2A target gene Xirp2 in Ang II-mediated cardiac remodeling was investigated. METHODS AND RESULTS: Here we demonstrate that the MEF2A target gene Xirp2 (also known as cardiomyopathy associated gene 3 [CMYA3]) is an important effector of the Ang II signaling pathway in the heart. Xirp2 belongs to the evolutionarily conserved, muscle-specific, actin-binding Xin gene family and is significantly induced in the heart in response to systemic administration of Ang II. Initially, we characterized the Xirp2 promoter and demonstrate that Ang II activates Xirp2 expression by stimulating MEF2A transcriptional activity. To further characterize the role of Xirp2 downstream of Ang II signaling we generated mice harboring a hypomorphic allele of the Xirp2 gene that resulted in a marked reduction in its expression in the heart. In the absence of Ang II, adult Xirp2 hypomorphic mice displayed cardiac hypertrophy and increased beta myosin heavy chain expression. Strikingly, Xirp2 hypomorphic mice chronically infused with Ang II exhibited altered pathological cardiac remodeling including an attenuated hypertrophic response, as well as diminished fibrosis and apoptosis. CONCLUSIONS: These findings reveal a novel MEF2A-Xirp2 pathway that functions downstream of Ang II signaling to modulate its pathological effects in the heart.


Asunto(s)
Cardiomegalia/metabolismo , Proteínas de Unión al ADN/metabolismo , Miocardio/metabolismo , Factores Reguladores Miogénicos/metabolismo , Proteínas Nucleares/metabolismo , Remodelación Ventricular , Angiotensina II/administración & dosificación , Animales , Apoptosis , Sitios de Unión , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Proteínas del Citoesqueleto , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Fibrosis , Regulación de la Expresión Génica , Bombas de Infusión Implantables , Infusiones Subcutáneas , Proteínas con Dominio LIM , Factores de Transcripción MEF2 , Ratones , Ratones Transgénicos , Miocardio/patología , Factores Reguladores Miogénicos/genética , Cadenas Pesadas de Miosina/metabolismo , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Transducción de Señal , Activación Transcripcional , Miosinas Ventriculares/metabolismo , Remodelación Ventricular/genética
14.
Nat Commun ; 13(1): 2185, 2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35449169

RESUMEN

Cardiac excitation-contraction coupling requires dyads, the nanoscopic microdomains formed adjacent to Z-lines by apposition of transverse tubules and junctional sarcoplasmic reticulum. Disruption of dyad architecture and function are common features of diseased cardiomyocytes. However, little is known about the mechanisms that modulate dyad organization during cardiac development, homeostasis, and disease. Here, we use proximity proteomics in intact, living hearts to identify proteins enriched near dyads. Among these proteins is CMYA5, an under-studied striated muscle protein that co-localizes with Z-lines, junctional sarcoplasmic reticulum proteins, and transverse tubules in mature cardiomyocytes. During cardiac development, CMYA5 positioning adjacent to Z-lines precedes junctional sarcoplasmic reticulum positioning or transverse tubule formation. CMYA5 ablation disrupts dyad architecture, dyad positioning at Z-lines, and junctional sarcoplasmic reticulum Ca2+ release, leading to cardiac dysfunction and inability to tolerate pressure overload. These data provide mechanistic insights into cardiomyopathy pathogenesis by demonstrating that CMYA5 anchors junctional sarcoplasmic reticulum to Z-lines, establishes dyad architecture, and regulates dyad Ca2+ release.


Asunto(s)
Acoplamiento Excitación-Contracción , Retículo Sarcoplasmático , Calcio/metabolismo , Proteínas Musculares/metabolismo , Miocitos Cardíacos/metabolismo , Sarcolema/metabolismo , Retículo Sarcoplasmático/metabolismo
15.
Nat Med ; 8(11): 1303-9, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12379849

RESUMEN

The four MEF2 transcription factors (MEF2A, -B, -C, and -D) regulate differentiation and calcium-dependent gene expression in muscle cells. We generated mice deficient in MEF2A, the predominant Mef2 gene product expressed in post-natal cardiac muscle. Most mice lacking Mef2a died suddenly within the first week of life and exhibited pronounced dilation of the right ventricle, myofibrillar fragmentation, mitochondrial disorganization and activation of a fetal cardiac gene program. The few Mef2a(-/-) mice that survived to adulthood also showed a deficiency of cardiac mitochondria and susceptibility to sudden death. Paradoxically, MEF2 transcriptional activity, revealed by the expression of a MEF2-dependent transgene, was enhanced in the hearts of Mef2a-mutant mice, reflecting the transcriptional activation of residual MEF2D. These findings reveal specific roles for MEF2A in maintaining appropriate mitochondrial content and cyto-architectural integrity in the post-natal heart and show that other MEF2 isoforms cannot support these activities.


Asunto(s)
Proteínas de Unión al ADN/genética , Muerte Súbita Cardíaca , Mitocondrias Cardíacas/fisiología , Factores de Transcripción/genética , Animales , Ensayo de Cambio de Movilidad Electroforética , Factores de Transcripción MEF2 , Ratones , Ratones Noqueados , Ratones Mutantes , Factores Reguladores Miogénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
J Cardiovasc Dev Dis ; 8(8)2021 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-34436226

RESUMEN

Non-coding RNAs (ncRNAs) play a key role in the regulation of transcriptional and epigenetic activity in mammalian cells. Comprehensive analysis of these ncRNAs has revealed sophisticated gene regulatory mechanisms which finely tune the proper gene output required for cellular homeostasis, proliferation, and differentiation. However, this elaborate circuitry has also made it vulnerable to perturbations that often result in disease. Among the many types of ncRNAs, long non-coding RNAs (lncRNAs) appear to have the most diverse mechanisms of action including competitive binding to miRNA targets, direct binding to mRNA, interactions with transcription factors, and facilitation of epigenetic modifications. Moreover, many lncRNAs display tissue-specific expression patterns suggesting an important regulatory role in organogenesis, yet the molecular mechanisms through which these molecules regulate cardiac and skeletal muscle development remains surprisingly limited. Given the structural and metabolic similarities of cardiac and skeletal muscle, it is likely that several lncRNAs expressed in both of these tissues have conserved functions in establishing the striated muscle phenotype. As many aspects of regeneration recapitulate development, understanding the role lncRNAs play in these processes may provide novel insights to improve regenerative therapeutic interventions in cardiac and skeletal muscle diseases. This review highlights key lncRNAs that function as regulators of development, regeneration, and disease in cardiac and skeletal muscle. Finally, we highlight lncRNAs encoded by imprinted genes in striated muscle and the contributions of these loci on the regulation of gene expression.

17.
Exp Neurobiol ; 29(3): 189-206, 2020 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-32606250

RESUMEN

Neurogenic differentiation 1 (NeuroD1) is a class B basic helix-loop-helix (bHLH) transcription factor and regulates differentiation and survival of neuronal and endocrine cells by means of several protein kinases, including extracellular signal-regulated kinase (ERK). However, the effect of phosphorylation on the functions of NeuroD1 by ERK has sparked controversy based on context-dependent differences across diverse species and cell types. Here, we evidenced that ERK-dependent phosphorylation controlled the stability of NeuroD1 and consequently, regulated proneural activity in neuronal cells. A null mutation at the ERK-dependent phosphorylation site, S274A, increased the half-life of NeuroD1 by blocking its ubiquitin-dependent proteasomal degradation. The S274A mutation did not interfere with either the nuclear translocation of NeuroD1 or its heterodimerization with E47, its ubiquitous partner and class A bHLH transcription factor. However, the S274A mutant increased transactivation of the E-box-mediated gene and neurite outgrowth in F11 neuroblastoma cells, compared to the wild-type NeuroD1. Transcriptome and Gene Ontology enrichment analyses indicated that genes involved in axonogenesis and dendrite development were downregulated in NeuroD1 knockout (KO) mice. Overexpression of the S274A mutant salvaged neurite outgrowth in NeuroD1-deficient mice, whereas neurite outgrowth was minimal with S274D, a phosphomimicking mutant. Our data indicated that a longer protein half-life enhanced the overall activity of NeuroD1 in stimulating downstream genes and neuronal differentiation. We propose that blocking ubiquitin-dependent proteasomal degradation may serve as a strategy to promote neuronal activity by stimulating the expression of neuron-specific genes in differentiating neurons.

18.
J Psychiatr Res ; 111: 8-15, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30658136

RESUMEN

CMYA5 is a candidate gene for schizophrenia because of the genetic association of variant rs10043986 (C > T) to this severe mental disorder. Studies of CMYA5 and its gene product, myospryn, in the brain and neuronal cells have not been previously reported. The SNP rs10043986 changes the 4,063rd amino acid from Pro to Leu, which is likely to alter protein function. To understand its potential role in the brain, we examined the neuronal expression of myospryn and its binding partner, desmin, an intermediate filament (IF) protein, and investigated how the two alleles of myospryn affect its binding to desmin. Myospryn and desmin are shown to be expressed in the brain and myospryn is shown to localize to the cytoplasm and nucleus of myoblast, neuroblastoma, and glioblastoma cell lines. Peripherin and vimentin, known brain IF proteins, have high protein similarity to desmin but were found not to interact with myospryn using yeast two-hybrid (Y2H). Using a quantitative Y2H assay and surface plasmon resonance, the T allele (Leu) of rs10043986 was found to have stronger binding to desmin than the C allele (Pro). Based on findings described in this report, we hypothesize that the interaction between myospryn to IF provides structural support and efficient rearrangement of the cytoskeleton network during early neuritogenesis.


Asunto(s)
Encéfalo/metabolismo , Desmina/metabolismo , Filamentos Intermedios/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Esquizofrenia/genética , Esquizofrenia/metabolismo , Alelos , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Periferinas/metabolismo , Polimorfismo de Nucleótido Simple , Unión Proteica , Resonancia por Plasmón de Superficie , Vimentina/metabolismo , Levaduras
19.
Biochim Biophys Acta ; 1773(6): 891-902, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17499862

RESUMEN

Recently we identified a novel target gene of MEF2A named myospryn that encodes a large, muscle-specific, costamere-restricted alpha-actinin binding protein. Myospryn belongs to the tripartite motif (TRIM) superfamily of proteins and was independently identified as a dysbindin-interacting protein. Dysbindin is associated with alpha-dystrobrevin, a component of the dystrophin-glycoprotein complex (DGC) in muscle. Apart from these initial findings little else is known regarding the potential function of myospryn in striated muscle. Here we reveal that myospryn is an anchoring protein for protein kinase A (PKA) (or AKAP) whose closest homolog is AKAP12, also known as gravin/AKAP250/SSeCKS. We demonstrate that myospryn co-localizes with RII alpha, a type II regulatory subunit of PKA, at the peripheral Z-disc/costameric region in striated muscle. Myospryn interacts with RII alpha and this scaffolding function has been evolutionarily conserved as the zebrafish ortholog also interacts with PKA. Moreover, myospryn serves as a substrate for PKA. These findings point to localized PKA signaling at the muscle costamere.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Transducción de Señal/fisiología , Proteínas de Anclaje a la Quinasa A , Secuencias de Aminoácidos/genética , Animales , Células COS , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Chlorocebus aethiops , Subunidad RIIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteína Quinasa Tipo II Dependiente de AMP Cíclico , Péptidos y Proteínas de Señalización Intracelular , Factores de Transcripción MEF2 , Ratones , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Proteínas Musculares/genética , Músculo Esquelético/citología , Factores Reguladores Miogénicos/metabolismo , Mapeo Peptídico , Homología de Secuencia de Aminoácido
20.
J Cardiovasc Dev Dis ; 5(3)2018 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-29996488

RESUMEN

The imprinted Dlk1-Dio3 genomic region harbors a noncoding RNA cluster encoding over fifty microRNAs (miRNAs), three long noncoding RNAs (lncRNAs), and a small nucleolar RNA (snoRNA) gene array. These distinct noncoding RNAs (ncRNAs) are thought to arise from a single polycistronic transcript that is subsequently processed into individual ncRNAs, each with important roles in diverse cellular contexts. Considering these ncRNAs are derived from a polycistron, it is possible that some coordinately regulate discrete biological processes in the heart. Here, we provide a comprehensive summary of Dlk1-Dio3 miRNAs and lncRNAs, as they are currently understood in the cellular and organ-level context of the cardiovascular system. Highlighted are expression profiles, mechanistic contributions, and functional roles of these ncRNAs in heart development and disease. Notably, a number of these ncRNAs are implicated in processes often perturbed in heart disease, including proliferation, differentiation, cell death, and fibrosis. However, most literature falls short of characterizing precise mechanisms for many of these ncRNAs, warranting further investigation. Taken together, the Dlk1-Dio3 locus represents a largely unexplored noncoding regulator of cardiac homeostasis, harboring numerous ncRNAs that may serve as therapeutic targets for cardiovascular disease.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA