Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Genes Dev ; 35(11-12): 835-840, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33985971

RESUMEN

Myocardin, a potent coactivator of serum response factor (SRF), competes with ternary complex factor (TCF) proteins for SRF binding to balance opposing mitogenic and myogenic gene programs in cardiac and smooth muscle. Here we identify a cardiac lncRNA transcribed adjacent to myocardin, named CARDINAL, which antagonizes SRF-dependent mitogenic gene transcription in the heart. CARDINAL-deficient mice show ectopic TCF/SRF-dependent mitogenic gene expression and decreased cardiac contractility in response to age and ischemic stress. CARDINAL forms a nuclear complex with SRF and inhibits TCF-mediated transactivation of the promitogenic gene c-fos, suggesting CARDINAL functions as an RNA cofactor for SRF in the heart.


Asunto(s)
Regulación de la Expresión Génica/genética , Corazón/fisiología , Proteínas Nucleares/metabolismo , ARN Largo no Codificante/metabolismo , Factor de Respuesta Sérica/metabolismo , Transactivadores/metabolismo , Factores de Edad , Animales , Modelos Animales de Enfermedad , Eliminación de Gen , Factores de Transcripción MEF2/metabolismo , Ratones , Ratones Endogámicos C57BL , Contracción Miocárdica/genética , Infarto del Miocardio/genética , Infarto del Miocardio/fisiopatología , Proteínas Nucleares/genética , ARN Largo no Codificante/genética , Factor de Respuesta Sérica/genética , Transactivadores/genética , Activación Transcripcional
2.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35101990

RESUMEN

Emerging evidence indicates that a subset of RNA molecules annotated as noncoding contain short open reading frames that code for small functional proteins called microproteins, which have largely been overlooked due to their small size. To search for cardiac-expressed microproteins, we used a comparative genomics approach and identified mitolamban (Mtlbn) as a highly conserved 47-amino acid transmembrane protein that is abundantly expressed in the heart. Mtlbn localizes specifically to the inner mitochondrial membrane where it interacts with subunits of complex III of the electron transport chain and with mitochondrial respiratory supercomplexes. Genetic deletion of Mtlbn in mice altered complex III assembly dynamics and reduced complex III activity. Unbiased metabolomic analysis of heart tissue from Mtlbn knockout mice further revealed an altered metabolite profile consistent with deficiencies in complex III activity. Cardiac-specific Mtlbn overexpression in transgenic (TG) mice induced cardiomyopathy with histological, biochemical, and ultrastructural pathologic features that contributed to premature death. Metabolomic analysis and biochemical studies indicated that hearts from Mtlbn TG mice exhibited increased oxidative stress and mitochondrial dysfunction. These findings reveal Mtlbn as a cardiac-expressed inner mitochondrial membrane microprotein that contributes to mitochondrial electron transport chain activity through direct association with complex III and the regulation of its assembly and function.


Asunto(s)
Cardiomiopatías/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/metabolismo , Miocardio/metabolismo , Animales , Cardiomiopatías/genética , Células Cultivadas , Complejo III de Transporte de Electrones/genética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/genética , Proteínas Mitocondriales/genética , Especificidad de Órganos
3.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34088848

RESUMEN

Homeothermic vertebrates produce heat in cold environments through thermogenesis, in which brown adipose tissue (BAT) increases mitochondrial oxidation along with uncoupling of the electron transport chain and activation of uncoupling protein 1 (UCP1). Although the transcription factors regulating the expression of UCP1 and nutrient oxidation genes have been extensively studied, only a few other proteins essential for BAT function have been identified. We describe the discovery of FAM195A, a BAT-enriched RNA binding protein, which is required for cold-dependent thermogenesis in mice. FAM195A knockout (KO) mice display whitening of BAT and an inability to thermoregulate. In BAT of FAM195A KO mice, enzymes involved in branched-chain amino acid (BCAA) metabolism are down-regulated, impairing their response to cold. Knockdown of FAM195A in brown adipocytes in vitro also impairs expression of leucine oxidation enzymes, revealing FAM195A to be a regulator of BCAA metabolism and a potential target for metabolic disorders.


Asunto(s)
Adipocitos Marrones , Tejido Adiposo Pardo , Frío , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Termogénesis , Aminoácidos de Cadena Ramificada/genética , Aminoácidos de Cadena Ramificada/metabolismo , Animales , Línea Celular Transformada , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados
4.
Proc Natl Acad Sci U S A ; 115(36): E8430-E8439, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30127033

RESUMEN

Twist transcription factors function as ancestral regulators of mesodermal cell fates in organisms ranging from Drosophila to mammals. Through lineage tracing of Twist2 (Tw2)-expressing cells with tamoxifen-inducible Tw2-CreERT2 and tdTomato (tdTO) reporter mice, we discovered a unique cell population that progressively contributes to cardiomyocytes (CMs), endothelial cells, and fibroblasts in the adult heart. Clonal analysis confirmed the ability of Tw2-derived tdTO+ (Tw2-tdTO+) cells to form CMs in vitro. Within the adult heart, Tw2-tdTO+ CMs accounted for ∼13% of total CMs, the majority of which resulted from fusion of Tw2-tdTO+ cells with existing CMs. Tw2-tdTO+ cells also contribute to cardiac remodeling after injury. We conclude that Tw2-tdTO+ cells participate in lifelong maintenance of cardiac function, at least in part through de novo formation of CMs and fusion with preexisting CMs, as well as in the genesis of other cellular components of the adult heart.


Asunto(s)
Células Madre Multipotentes/metabolismo , Miocardio/metabolismo , Proteínas Represoras/biosíntesis , Proteína 1 Relacionada con Twist/biosíntesis , Animales , Drosophila melanogaster , Células Endoteliales/citología , Células Endoteliales/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Ratones , Ratones Transgénicos , Células Madre Multipotentes/citología , Miocardio/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteínas Represoras/genética , Proteína 1 Relacionada con Twist/genética
5.
J Biol Chem ; 293(23): 8802-8811, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29653945

RESUMEN

Control of protein homeostasis is an essential cellular process that, when perturbed, can result in the deregulation or toxic accumulation of proteins. Owing to constant mechanical stress, striated muscle proteins are particularly prone to wear and tear and require several protein quality-control mechanisms to coordinate protein turnover and removal of damaged proteins. Kelch-like proteins, substrate adapters for the Cullin-3 (Cul3)-RING ligase (CRL3) complex, are emerging as critical regulators of striated muscle development and function, highlighting the importance of Cul3-mediated proteostasis in muscle function. To explore the role of Cul3-mediated proteostasis in striated muscle, here we deleted Cul3 specifically in either skeletal muscle (SkM-Cul3 KO) or cardiomyocytes (CM-Cul3 KO) of mice. The loss of Cul3 caused neonatal lethality and dramatic alterations in the proteome, which were unique to each striated muscle type. Many of the proteins whose expression was significantly changed in the SkM-Cul3 KO were components of the extracellular matrix and sarcomere, whereas proteins altered in the CM-Cul3 KO were involved in metabolism. These findings highlight the requirement for striated muscle-specific CRL3 activity and indicate how the CRL3 complex can control different nodes of protein interaction networks in different types of striated muscle. Further identification of Cul3 substrates, and how these substrates are targeted, may reveal therapeutic targets and treatment regimens for striated muscle diseases.


Asunto(s)
Proteínas Cullin/genética , Eliminación de Gen , Músculo Estriado/patología , Miocitos Cardíacos/patología , Animales , Células Cultivadas , Proteínas Cullin/metabolismo , Regulación del Desarrollo de la Expresión Génica , Metaboloma , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Estriado/embriología , Músculo Estriado/metabolismo , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Mapas de Interacción de Proteínas
6.
Development ; 143(15): 2853-61, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27385017

RESUMEN

Myocardin-related transcription factors (MRTFs) play a central role in the regulation of actin expression and cytoskeletal dynamics. Stimuli that promote actin polymerization allow for shuttling of MRTFs to the nucleus where they activate serum response factor (SRF), a regulator of actin and other cytoskeletal protein genes. SRF is an essential regulator of skeletal muscle differentiation and numerous components of the muscle sarcomere, but the potential involvement of MRTFs in skeletal muscle development has not been examined. We explored the role of MRTFs in muscle development in vivo by generating mutant mice harboring a skeletal muscle-specific deletion of MRTF-B and a global deletion of MRTF-A. These double knockout (dKO) mice were able to form sarcomeres during embryogenesis. However, the sarcomeres were abnormally small and disorganized, causing skeletal muscle hypoplasia and perinatal lethality. Transcriptome analysis demonstrated dramatic dysregulation of actin genes in MRTF dKO mice, highlighting the importance of MRTFs in actin cycling and myofibrillogenesis. MRTFs were also shown to be necessary for the survival of skeletal myoblasts and for the efficient formation of intact myotubes. Our findings reveal a central role for MRTFs in sarcomere formation during skeletal muscle development and point to the potential involvement of these transcriptional co-activators in skeletal myopathies.


Asunto(s)
Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Actinas/metabolismo , Animales , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Microscopía Electrónica , Desarrollo de Músculos/fisiología , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Transactivadores/genética , Factores de Transcripción/genética
7.
Nature ; 499(7458): 301-5, 2013 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-23868259

RESUMEN

Fusion of myoblasts is essential for the formation of multi-nucleated muscle fibres. However, the identity of muscle-specific proteins that directly govern this fusion process in mammals has remained elusive. Here we identify a muscle-specific membrane protein, named myomaker, that controls myoblast fusion. Myomaker is expressed on the cell surface of myoblasts during fusion and is downregulated thereafter. Overexpression of myomaker in myoblasts markedly enhances fusion, and genetic disruption of myomaker in mice causes perinatal death due to an absence of multi-nucleated muscle fibres. Remarkably, forced expression of myomaker in fibroblasts promotes fusion with myoblasts, demonstrating the direct participation of this protein in the fusion process. Pharmacological perturbation of the actin cytoskeleton abolishes the activity of myomaker, consistent with previous studies implicating actin dynamics in myoblast fusion. These findings reveal a long-sought myogenic fusion protein that controls mammalian myoblast fusion and provide new insights into the molecular underpinnings of muscle formation.


Asunto(s)
Proteínas de la Membrana/fisiología , Desarrollo de Músculos , Proteínas Musculares/fisiología , Músculo Esquelético/embriología , Mioblastos/citología , Animales , Fusión Celular , Línea Celular , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos/metabolismo
8.
Proc Natl Acad Sci U S A ; 113(31): E4494-503, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27418600

RESUMEN

Innervation of skeletal muscle by motor neurons occurs through the neuromuscular junction, a cholinergic synapse essential for normal muscle growth and function. Defects in nerve-muscle signaling cause a variety of neuromuscular disorders with features of ataxia, paralysis, skeletal muscle wasting, and degeneration. Here we show that the nuclear zinc finger protein ZFP106 is highly enriched in skeletal muscle and is required for postnatal maintenance of myofiber innervation by motor neurons. Genetic disruption of Zfp106 in mice results in progressive ataxia and hindlimb paralysis associated with motor neuron degeneration, severe muscle wasting, and premature death by 6 mo of age. We show that ZFP106 is an RNA-binding protein that associates with the core splicing factor RNA binding motif protein 39 (RBM39) and localizes to nuclear speckles adjacent to spliceosomes. Upon inhibition of pre-mRNA synthesis, ZFP106 translocates with other splicing factors to the nucleolus. Muscle and spinal cord of Zfp106 knockout mice displayed a gene expression signature of neuromuscular degeneration. Strikingly, altered splicing of the Nogo (Rtn4) gene locus in skeletal muscle of Zfp106 knockout mice resulted in ectopic expression of NOGO-A, the neurite outgrowth factor that inhibits nerve regeneration and destabilizes neuromuscular junctions. These findings reveal a central role for Zfp106 in the maintenance of nerve-muscle signaling, and highlight the involvement of aberrant RNA processing in neuromuscular disease pathogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Síndrome Debilitante/genética , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Células COS , Chlorocebus aethiops , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Desnervación Muscular , Músculo Esquelético/inervación , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Síndrome Debilitante/metabolismo
10.
Proc Natl Acad Sci U S A ; 111(11): 4109-14, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24591619

RESUMEN

Regeneration of adult skeletal muscle following injury occurs through the activation of satellite cells, an injury-sensitive muscle stem cell population that proliferates, differentiates, and fuses with injured myofibers. Members of the myocyte enhancer factor 2 (MEF2) family of transcription factors play essential roles in muscle differentiation during embryogenesis, but their potential contributions to adult muscle regeneration have not been systematically explored. To investigate the potential involvement of MEF2 factors in muscle regeneration, we conditionally deleted the Mef2a, c, and d genes, singly and in combination, within satellite cells in mice, using tamoxifen-inducible Cre recombinase under control of the satellite cell-specific Pax7 promoter. We show that deletion of individual Mef2 genes has no effect on muscle regeneration in response to cardiotoxin injury. However, combined deletion of the Mef2a, c, and d genes results in a blockade to regeneration. Satellite cell-derived myoblasts lacking MEF2A, C, and D proliferate normally in culture, but cannot differentiate. The absence of MEF2A, C, and D in satellite cells is associated with aberrant expression of a broad collection of known and unique protein-coding and long noncoding RNA genes. These findings reveal essential and redundant roles of MEF2A, C, and D in satellite cell differentiation and identify a MEF2-dependent transcriptome associated with skeletal muscle regeneration.


Asunto(s)
Regulación de la Expresión Génica/genética , Músculo Esquelético/crecimiento & desarrollo , Regeneración/fisiología , Células Satélite del Músculo Esquelético/metabolismo , Animales , Diferenciación Celular/genética , Citometría de Flujo , Inmunohistoquímica , Factores de Transcripción MEF2/deficiencia , Factores de Transcripción MEF2/metabolismo , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Cardiovasc Res ; 120(1): 56-68, 2024 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-37890031

RESUMEN

AIMS: RNA binding proteins play essential roles in mediating RNA splicing and are key post-transcriptional regulators in the heart. Our recent study demonstrated that RBPMS (RNA binding protein with multiple splicing) is crucial for cardiac development through modulating mRNA splicing, but little is known about its functions in the adult heart. In this study, we aim to characterize the post-natal cardiac function of Rbpms and its mechanism of action. METHODS AND RESULTS: We generated a cardiac-specific knockout mouse line and found that cardiac-specific loss of Rbpms caused severe cardiomyocyte contractile defects, leading to dilated cardiomyopathy and early lethality in adult mice. We showed by proximity-dependent biotin identification assay and mass spectrometry that RBPMS associates with spliceosome factors and other RNA binding proteins, such as RBM20, that are important in cardiac function. We performed paired-end RNA sequencing and RT-PCR and found that RBPMS regulates mRNA alternative splicing of genes associated with sarcomere structure and function, such as Ttn, Pdlim5, and Nexn, generating new protein isoforms. Using a minigene splicing reporter assay, we determined that RBPMS regulates target gene splicing through recognizing tandem intronic CAC motifs. We also showed that RBPMS knockdown in human induced pluripotent stem cell-derived cardiomyocytes impaired cardiomyocyte contraction. CONCLUSION: This study identifies RBPMS as an important regulator of cardiomyocyte contraction and cardiac function by modulating sarcomeric gene alternative splicing.


Asunto(s)
Empalme Alternativo , Células Madre Pluripotentes Inducidas , Animales , Humanos , Ratones , Conectina/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Ratones Noqueados , Miocitos Cardíacos/metabolismo , ARN/metabolismo , Empalme del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
12.
Sci Adv ; 10(11): eadk7160, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38489358

RESUMEN

During development, cells make switch-like decisions to activate new gene programs specifying cell lineage. The mechanisms underlying these decisive choices remain unclear. Here, we show that the cardiovascular transcriptional coactivator myocardin (MYOCD) activates cell identity genes by concentration-dependent and switch-like formation of transcriptional condensates. MYOCD forms such condensates and activates cell identity genes at critical concentration thresholds achieved during smooth muscle cell and cardiomyocyte differentiation. The carboxyl-terminal disordered region of MYOCD is necessary and sufficient for condensate formation. Disrupting this region's ability to form condensates disrupts gene activation and smooth muscle cell reprogramming. Rescuing condensate formation by replacing this region with disordered regions from functionally unrelated proteins rescues gene activation and smooth muscle cell reprogramming. Our findings demonstrate that MYOCD condensate formation is required for gene activation during cardiovascular differentiation. We propose that the formation of transcriptional condensates at critical concentrations of cell type-specific regulators provides a molecular switch underlying the activation of key cell identity genes during development.


Asunto(s)
Miocitos del Músculo Liso , Factores de Transcripción , Linaje de la Célula/genética , Diferenciación Celular/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Miocitos del Músculo Liso/metabolismo , Activación Transcripcional
13.
Dev Cell ; 58(24): 2867-2880.e7, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-37972593

RESUMEN

Cardiomyocytes are highly metabolic cells responsible for generating the contractile force in the heart. During fetal development and regeneration, these cells actively divide but lose their proliferative activity in adulthood. The mechanisms that coordinate their metabolism and proliferation are not fully understood. Here, we study the role of the transcription factor NFYa in developing mouse hearts. Loss of NFYa alters cardiomyocyte composition, causing a decrease in immature regenerative cells and an increase in trabecular and mature cardiomyocytes, as identified by spatial and single-cell transcriptome analyses. NFYa-deleted cardiomyocytes exhibited reduced proliferation and impaired mitochondrial metabolism, leading to cardiac growth defects and embryonic death. NFYa, interacting with cofactor SP2, activates genes linking metabolism and proliferation at the transcription level. Our study identifies a nodal role of NFYa in regulating prenatal cardiac growth and a previously unrecognized transcriptional control mechanism of heart metabolism, highlighting the importance of mitochondrial metabolism during heart development and regeneration.


Asunto(s)
Miocitos Cardíacos , Factores de Transcripción , Animales , Ratones , Proliferación Celular/fisiología , Desarrollo Fetal , Corazón Fetal/metabolismo , Corazón/fisiología , Miocitos Cardíacos/metabolismo , Factores de Transcripción/metabolismo
14.
Nat Commun ; 14(1): 4333, 2023 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-37468485

RESUMEN

Skeletal muscle fibers express distinct gene programs during development and maturation, but the underlying gene regulatory networks that confer stage-specific myofiber properties remain unknown. To decipher these distinctive gene programs and how they respond to neural activity, we generated a combined multi-omic single-nucleus RNA-seq and ATAC-seq atlas of mouse skeletal muscle development at multiple stages of embryonic, fetal, and postnatal life. We found that Myogenin, Klf5, and Tead4 form a transcriptional complex that synergistically activates the expression of muscle genes in developing myofibers. During myofiber maturation, the transcription factor Maf acts as a transcriptional switch to activate the mature fast muscle gene program. In skeletal muscles of mutant mice lacking voltage-gated L-type Ca2+ channels (Cav1.1), Maf expression and myofiber maturation are impaired. These findings provide a transcriptional atlas of muscle development and reveal genetic links between myofiber formation, maturation, and contraction.


Asunto(s)
Fibras Musculares Esqueléticas , Músculo Esquelético , Ratones , Animales , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Regulación de la Expresión Génica , Factores de Transcripción/metabolismo , Diferenciación Celular
15.
J Cell Biol ; 179(3): 527-37, 2007 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-17984325

RESUMEN

Atrial cardiomyocytes, neurons, and endocrine tissues secrete neurotransmitters and peptide hormones via large dense-core vesicles (LDCVs). We describe a new member of the Ras family of G-proteins, named RRP17, which is expressed specifically in cardiomyocytes, neurons, and the pancreas. RRP17 interacts with Ca(2+)-activated protein for secretion-1 (CAPS1), one of only a few proteins known to be associated exclusively with LDCV exocytosis. Ectopic expression of RRP17 in cardiomyocytes enhances secretion of atrial natriuretic peptide (ANP), a regulator of blood pressure and natriuresis. Conversely, genetic deletion of RRP17 in mice results in dysmorphic LDCVs, impaired ANP secretion, and hypertension. These findings identify RRP17 as a component of the cellular machinery involved in regulated secretion within the heart and potential mediator of the endocrine influence of the heart on other tissues.


Asunto(s)
Factor Natriurético Atrial/metabolismo , Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica , Proteínas ras/metabolismo , Secuencia de Aminoácidos , Animales , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/fisiología , Proteínas de Unión al Calcio/metabolismo , Atrios Cardíacos/metabolismo , Humanos , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Miocitos Cardíacos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Páncreas/metabolismo , Homología de Secuencia de Aminoácido
16.
Nat Cell Biol ; 23(5): 467-475, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33941892

RESUMEN

Direct cardiac reprogramming of fibroblasts to cardiomyocytes presents an attractive therapeutic strategy to restore cardiac function following injury. Cardiac reprogramming was initially achieved through overexpression of the transcription factors Gata4, Mef2c and Tbx5; later, Hand2 and Akt1 were found to further enhance this process1-5. Yet, staunch epigenetic barriers severely limit the ability of these cocktails to reprogramme adult fibroblasts6,7. We undertook a screen of mammalian gene regulatory factors to discover novel regulators of cardiac reprogramming in adult fibroblasts and identified the histone reader PHF7 as the most potent activating factor8. Mechanistically, PHF7 localizes to cardiac super enhancers in fibroblasts, and through cooperation with the SWI/SNF complex, it increases chromatin accessibility and transcription factor binding at these sites. Furthermore, PHF7 recruits cardiac transcription factors to activate a positive transcriptional autoregulatory circuit in reprogramming. Importantly, PHF7 achieves efficient reprogramming in the absence of Gata4. Here, we highlight the underexplored necessity of cardiac epigenetic readers, such as PHF7, in harnessing chromatin remodelling and transcriptional complexes to overcome critical barriers to direct cardiac reprogramming.


Asunto(s)
Factor de Transcripción GATA4/metabolismo , Histonas/metabolismo , Transducción de Señal/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Reprogramación Celular , Fibroblastos/metabolismo , Factor de Transcripción GATA4/genética , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/fisiología , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/genética
17.
Proc Natl Acad Sci U S A ; 104(52): 20844-9, 2007 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-18093911

RESUMEN

The muscle-specific microRNAs, miR-1 and miR-133, play important roles in muscle growth and differentiation. Here, we show that the MEF2 transcription factor, an essential regulator of muscle development, directly activates transcription of a bicistronic primary transcript encoding miR-1-2 and 133a-1 via an intragenic muscle-specific enhancer located between the miR-1-2 and 133a-1 coding regions. This MEF2-dependent enhancer is activated in the linear heart tube during mouse embryogenesis and thereafter controls transcription throughout the atrial and ventricular chambers of the heart. MEF2 together with MyoD also regulates the miR-1-2/-133a-1 intragenic enhancer in the somite myotomes and in all skeletal muscle fibers during embryogenesis and adulthood. A similar muscle-specific intragenic enhancer controls transcription of the miR-1-1/-133a-2 locus. These findings reveal a common architecture of regulatory elements associated with the miR-1/-133 genes and underscore the central role of MEF2 as a regulator of the transcriptional and posttranscriptional pathways that control cardiac and skeletal muscle development.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , MicroARNs/genética , Músculo Esquelético/embriología , Músculo Esquelético/metabolismo , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/fisiología , Transcripción Genética , Animales , Eliminación de Gen , Corazón/embriología , Factores de Transcripción MEF2 , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Genéticos , Miocardio/metabolismo
18.
J Clin Invest ; 116(7): 1853-64, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16767219

RESUMEN

Class IIa histone deacetylases (HDACs) regulate a variety of cellular processes, including cardiac growth, bone development, and specification of skeletal muscle fiber type. Multiple serine/threonine kinases control the subcellular localization of these HDACs by phosphorylation of common serine residues, but whether certain class IIa HDACs respond selectively to specific kinases has not been determined. Here we show that calcium/calmodulin-dependent kinase II (CaMKII) signals specifically to HDAC4 by binding to a unique docking site that is absent in other class IIa HDACs. Phosphorylation of HDAC4 by CaMKII promotes nuclear export and prevents nuclear import of HDAC4, with consequent derepression of HDAC target genes. In cardiomyocytes, CaMKII phosphorylation of HDAC4 results in hypertrophic growth, which can be blocked by a signal-resistant HDAC4 mutant. These findings reveal a central role for HDAC4 in CaMKII signaling pathways and have implications for the control of gene expression by calcium signaling in a variety of cell types.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Histona Desacetilasas/metabolismo , Isoenzimas/metabolismo , Miocitos Cardíacos , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Proteínas 14-3-3/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células COS , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Núcleo Celular/metabolismo , Chlorocebus aethiops , Activación Enzimática , Histona Desacetilasas/genética , Hipertrofia , Isoenzimas/genética , Datos de Secuencia Molecular , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/genética , Alineación de Secuencia , Técnicas del Sistema de Dos Híbridos
19.
Mol Biol Cell ; 17(1): 438-47, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16236793

RESUMEN

Class II histone deacetylases (HDACs) contain unique amino-terminal extensions that mediate interactions with members of the myocyte enhancer factor-2 (MEF2) family of transcription factors and responsiveness to kinases, including Ca2+/calmodulin-dependent kinase (CaMK). Despite intense investigation of class II HDACs, little is known of MEF2-independent mechanisms for transcriptional repression by these chromatin-modifying enzymes. Here, we demonstrate that class II HDACs 4 and 5 physically associate with ankyrin-repeat proteins ANKRA2 and RFXANK (RFX-B/Tvl-1/ANKRA1). ANKRA2 is a megalin- and BKCa potassium channel-interacting factor, whereas RFXANK is a positive regulator of major histocompatibility complex II (MHC II) gene expression. HDAC4 and HDAC5 interact with the ankyrin repeats of ANKRA2 and RFXANK and, through association with RFXANK, repress MHC II promoter activation. HDACs 4 and 5 also repress endogenous HLA-DRA gene expression induced by CIITA. Phosphorylation of class II HDACs by CaMK results in CRM1-dependent nuclear export of HDAC/RFXANK complexes. These results define a novel transcriptional pathway under the control of class II HDACs and suggest a role for these transcriptional repressors as signal-responsive regulators of antigen presentation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ancirinas/metabolismo , Histona Desacetilasas/metabolismo , Glicoproteínas de Membrana/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Ancirinas/genética , Línea Celular , Chlorocebus aethiops , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Histona Desacetilasas/clasificación , Histona Desacetilasas/genética , Macrófagos/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Regiones Promotoras Genéticas/genética , Unión Proteica , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Factores de Transcripción
20.
Elife ; 72018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30299255

RESUMEN

Calcium (Ca2+) dysregulation is a hallmark of heart failure and is characterized by impaired Ca2+ sequestration into the sarcoplasmic reticulum (SR) by the SR-Ca2+-ATPase (SERCA). We recently discovered a micropeptide named DWORF (DWarf Open Reading Frame) that enhances SERCA activity by displacing phospholamban (PLN), a potent SERCA inhibitor. Here we show that DWORF has a higher apparent binding affinity for SERCA than PLN and that DWORF overexpression mitigates the contractile dysfunction associated with PLN overexpression, substantiating its role as a potent activator of SERCA. Additionally, using a well-characterized mouse model of dilated cardiomyopathy (DCM) due to genetic deletion of the muscle-specific LIM domain protein (MLP), we show that DWORF overexpression restores cardiac function and prevents the pathological remodeling and Ca2+ dysregulation classically exhibited by MLP knockout mice. Our results establish DWORF as a potent activator of SERCA within the heart and as an attractive candidate for a heart failure therapeutic.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Cardiomiopatía Dilatada/fisiopatología , Contracción Miocárdica/efectos de los fármacos , Péptidos/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Animales , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Insuficiencia Cardíaca/prevención & control , Proteínas con Dominio LIM/deficiencia , Ratones Noqueados , Proteínas Musculares/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA