Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 818
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 162(2): 300-313, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26144318

RESUMEN

The transition from proliferating precursor cells to post-mitotic differentiated cells is crucial for development, tissue homeostasis, and tumor suppression. To study cell-cycle exit during differentiation in vivo, we developed a conditional knockout and lineage-tracing system for Caenorhabditis elegans. Combined lineage-specific gene inactivation and genetic screening revealed extensive redundancies between previously identified cell-cycle inhibitors and the SWI/SNF chromatin-remodeling complex. Muscle precursor cells missing either SWI/SNF or G1/S inhibitor function could still arrest cell division, while simultaneous inactivation of these regulators caused continued proliferation and a C. elegans tumor phenotype. Further genetic analyses support that SWI/SNF acts in concert with hlh-1 MyoD, antagonizes Polycomb-mediated transcriptional repression, and suppresses cye-1 Cyclin E transcription to arrest cell division of muscle precursors. Thus, SWI/SNF and G1/S inhibitors provide alternative mechanisms to arrest cell-cycle progression during terminal differentiation, which offers insight into the frequent mutation of SWI/SNF genes in human cancers.


Asunto(s)
Caenorhabditis elegans/citología , Caenorhabditis elegans/crecimiento & desarrollo , Músculos/citología , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Ciclo Celular , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Musculares , Músculos/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Factores Reguladores Miogénicos/metabolismo , Proteínas Nucleares , Proteínas del Grupo Polycomb/metabolismo , Factores de Transcripción/metabolismo
2.
Cell ; 155(2): 435-47, 2013 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-24075010

RESUMEN

Infections disturb metabolic homeostasis in many contexts, but the underlying connections are not completely understood. To address this, we use paired genetic and computational screens in Drosophila to identify transcriptional regulators of immunity and pathology and their associated target genes and physiologies. We show that Mef2 is required in the fat body for anabolic function and the immune response. Using genetic and biochemical approaches, we find that MEF2 is phosphorylated at a conserved site in healthy flies and promotes expression of lipogenic and glycogenic enzymes. Upon infection, this phosphorylation is lost, and the activity of MEF2 changes--MEF2 now associates with the TATA binding protein to bind a distinct TATA box sequence and promote antimicrobial peptide expression. The loss of phosphorylated MEF2 contributes to loss of anabolic enzyme expression in Gram-negative bacterial infection. MEF2 is thus a critical transcriptional switch in the adult fat body between metabolism and immunity.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/inmunología , Drosophila melanogaster/metabolismo , Factores Reguladores Miogénicos/metabolismo , Secuencia de Aminoácidos , Animales , Candida albicans , Proteínas de Drosophila/inmunología , Drosophila melanogaster/microbiología , Enterobacter cloacae , Cuerpo Adiposo/metabolismo , Regulación de la Expresión Génica , Glucógeno/metabolismo , Metabolismo , Mycobacterium marinum , Factores Reguladores Miogénicos/inmunología , Fosforilación , Proteína de Unión a TATA-Box/metabolismo
3.
Development ; 151(20)2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39114943

RESUMEN

Vertebrates and tunicates are sister groups that share a common fusogenic factor, Myomaker (Mymk), that drives myoblast fusion and muscle multinucleation. Yet they are divergent in when and where they express Mymk. In vertebrates, all developing skeletal muscles express Mymk and are obligately multinucleated. In tunicates, Mymk is expressed only in post-metamorphic multinucleated muscles, but is absent from mononucleated larval muscles. In this study, we demonstrate that cis-regulatory sequence differences in the promoter region of Mymk underlie the different spatiotemporal patterns of its transcriptional activation in tunicates and vertebrates. Although in vertebrates myogenic regulatory factors (MRFs) such as MyoD1 alone are required and sufficient for Mymk transcription in all skeletal muscles, we show that transcription of Mymk in post-metamorphic muscles of the tunicate Ciona requires the combinatorial activity of MRF, MyoD and Early B-cell Factor (Ebf). This macroevolutionary difference appears to be encoded in cis, likely due to the presence of a putative Ebf-binding site adjacent to predicted MRF binding sites in the Ciona Mymk promoter. We further discuss how Mymk and myoblast fusion might have been regulated in the last common ancestor of tunicates and vertebrates, for which we propose two models.


Asunto(s)
Regiones Promotoras Genéticas , Animales , Regiones Promotoras Genéticas/genética , Proteína MioD/metabolismo , Proteína MioD/genética , Regulación del Desarrollo de la Expresión Génica , Músculo Esquelético/metabolismo , Factores Reguladores Miogénicos/metabolismo , Factores Reguladores Miogénicos/genética , Urocordados/genética , Urocordados/embriología , Desarrollo de Músculos/genética
4.
Cell ; 151(1): 41-55, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-23021214

RESUMEN

Natural sensory input shapes both structure and function of developing neurons, but how early experience-driven morphological and physiological plasticity are interrelated remains unclear. Using rapid time-lapse two-photon calcium imaging of network activity and single-neuron growth within the unanesthetized developing brain, we demonstrate that visual stimulation induces coordinated changes to neuronal responses and dendritogenesis. Further, we identify the transcription factor MEF2A/2D as a major regulator of neuronal response to plasticity-inducing stimuli directing both structural and functional changes. Unpatterned sensory stimuli that change plasticity thresholds induce rapid degradation of MEF2A/2D through a classical apoptotic pathway requiring NMDA receptors and caspases-9 and -3/7. Knockdown of MEF2A/2D alone is sufficient to induce a metaplastic shift in threshold of both functional and morphological plasticity. These findings demonstrate how sensory experience acting through altered levels of the transcription factor MEF2 fine-tunes the plasticity thresholds of brain neurons during neural circuit formation.


Asunto(s)
Encéfalo/embriología , Factores Reguladores Miogénicos/metabolismo , Plasticidad Neuronal , Factores de Transcripción/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Animales , Percepción Auditiva , Encéfalo/citología , Caspasas/metabolismo , Factores de Transcripción MEF2 , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sonido , Percepción Visual
5.
Cell ; 151(7): 1581-94, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23260144

RESUMEN

The activity-dependent transcription factor myocyte enhancer factor 2 (MEF2) induces excitatory synapse elimination in mouse neurons, which requires fragile X mental retardation protein (FMRP), an RNA-binding protein implicated in human cognitive dysfunction and autism. We report here that protocadherin 10 (Pcdh10), an autism-spectrum disorders gene, is necessary for this process. MEF2 and FMRP cooperatively regulate the expression of Pcdh10. Upon MEF2 activation, PSD-95 is ubiquitinated by the ubiquitin E3 ligase murine double minute 2 (Mdm2) and then binds to Pcdh10, which links it to the proteasome for degradation. Blockade of the Pcdh10-proteasome interaction inhibits MEF2-induced PSD-95 degradation and synapse elimination. In FMRP-lacking neurons, elevated protein levels of eukaryotic translation elongation factor 1 α (EF1α), an Mdm2-interacting protein and FMRP target mRNA, sequester Mdm2 and prevent MEF2-induced PSD-95 ubiquitination and synapse elimination. Together, our findings reveal roles for multiple autism-linked genes in activity-dependent synapse elimination.


Asunto(s)
Guanilato-Quinasas/metabolismo , Hipocampo/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Animales , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Cadherinas/metabolismo , Dendritas/metabolismo , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Hipocampo/citología , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Protocadherinas , Sinapsis/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
6.
Nucleic Acids Res ; 52(5): 2711-2723, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38281192

RESUMEN

Class IIa Histone deacetylases (HDACs), including HDAC4, 5, 7 and 9, play key roles in multiple important developmental and differentiation processes. Recent studies have shown that class IIa HDACs exert their transcriptional repressive function by interacting with tissue-specific transcription factors, such as members of the myocyte enhancer factor 2 (MEF2) family of transcription factors. However, the molecular mechanism is not well understood. In this study, we determined the crystal structure of an HDAC4-MEF2A-DNA complex. This complex adopts a dumbbell-shaped overall architecture, with a 2:4:2 stoichiometry of HDAC4, MEF2A and DNA molecules. In the complex, two HDAC4 molecules form a dimer through the interaction of their glutamine-rich domain (GRD) to form the stem of the 'dumbbell'; while two MEF2A dimers and their cognate DNA molecules are bridged by the HDAC4 dimer. Our structural observations were then validated using biochemical and mutagenesis assays. Further cell-based luciferase reporter gene assays revealed that the dimerization of HDAC4 is crucial in its ability to repress the transcriptional activities of MEF2 proteins. Taken together, our findings not only provide the structural basis for the assembly of the HDAC4-MEF2A-DNA complex but also shed light on the molecular mechanism of HDAC4-mediated long-range gene regulation.


Asunto(s)
ADN , Histona Desacetilasas , Factores de Transcripción MEF2 , Proteínas Represoras , ADN/química , ADN/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Factores de Transcripción MEF2/química , Factores de Transcripción MEF2/metabolismo , Factores Reguladores Miogénicos/química , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Humanos , Histona Desacetilasas/química , Histona Desacetilasas/metabolismo
7.
Nat Immunol ; 14(10): 1084-92, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23974956

RESUMEN

MEF2B encodes a transcriptional activator and is mutated in ∼11% of diffuse large B cell lymphomas (DLBCLs) and ∼12% of follicular lymphomas (FLs). Here we found that MEF2B directly activated the transcription of the proto-oncogene BCL6 in normal germinal-center (GC) B cells and was required for DLBCL proliferation. Mutation of MEF2B resulted in enhanced transcriptional activity of MEF2B either through disruption of its interaction with the corepressor CABIN1 or by rendering it insensitive to inhibitory signaling events mediated by phosphorylation and sumoylation. Consequently, the transcriptional activity of Bcl-6 was deregulated in DLBCLs with MEF2B mutations. Thus, somatic mutations of MEF2B may contribute to lymphomagenesis by deregulating BCL6 expression, and MEF2B may represent an alternative target for blocking Bcl-6 activity in DLBCLs.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Linfoma de Células B Grandes Difuso/genética , Proteínas de Dominio MADS/genética , Mutación , Factores Reguladores Miogénicos/genética , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Ciclo Celular/genética , Proliferación Celular , Análisis por Conglomerados , Biología Computacional , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Perfilación de la Expresión Génica , Centro Germinal/metabolismo , Centro Germinal/patología , Humanos , Linfoma Folicular/genética , Linfoma Folicular/metabolismo , Linfoma de Células B Grandes Difuso/metabolismo , Proteínas de Dominio MADS/química , Proteínas de Dominio MADS/metabolismo , Factores de Transcripción MEF2 , Ratones , Simulación del Acoplamiento Molecular , Factores Reguladores Miogénicos/química , Factores Reguladores Miogénicos/metabolismo , Unión Proteica , Conformación Proteica , Proto-Oncogenes Mas , Sumoilación/genética , Transcripción Genética
8.
Cell ; 142(3): 375-86, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20691899

RESUMEN

The reprogramming of fibroblasts to induced pluripotent stem cells (iPSCs) raises the possibility that a somatic cell could be reprogrammed to an alternative differentiated fate without first becoming a stem/progenitor cell. A large pool of fibroblasts exists in the postnatal heart, yet no single "master regulator" of direct cardiac reprogramming has been identified. Here, we report that a combination of three developmental transcription factors (i.e., Gata4, Mef2c, and Tbx5) rapidly and efficiently reprogrammed postnatal cardiac or dermal fibroblasts directly into differentiated cardiomyocyte-like cells. Induced cardiomyocytes expressed cardiac-specific markers, had a global gene expression profile similar to cardiomyocytes, and contracted spontaneously. Fibroblasts transplanted into mouse hearts one day after transduction of the three factors also differentiated into cardiomyocyte-like cells. We believe these findings demonstrate that functional cardiomyocytes can be directly reprogrammed from differentiated somatic cells by defined factors. Reprogramming of endogenous or explanted fibroblasts might provide a source of cardiomyocytes for regenerative approaches.


Asunto(s)
Diferenciación Celular , Fibroblastos/citología , Miocardio/citología , Miocitos Cardíacos/citología , Animales , Separación Celular , Fibroblastos/metabolismo , Factor de Transcripción GATA4/metabolismo , Perfilación de la Expresión Génica , Factores de Transcripción MEF2 , Ratones , Contracción Muscular , Miocitos Cardíacos/metabolismo , Factores Reguladores Miogénicos/metabolismo , Proteínas de Dominio T Box/metabolismo
9.
Cell ; 140(4): 554-66, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20178747

RESUMEN

Skeletal myogenesis, like hematopoiesis, occurs in successive developmental stages that involve different cell populations and expression of different genes. We show here that the transcription factor nuclear factor one X (Nfix), whose expression is activated by Pax7 in fetal muscle, in turn activates the transcription of fetal specific genes such as MCK and beta-enolase while repressing embryonic genes such as slow myosin. In the case of the MCK promoter, Nfix forms a complex with PKC theta that binds, phosphorylates, and activates MEF2A. Premature expression of Nfix activates fetal and suppresses embryonic genes in embryonic muscle, whereas muscle-specific ablation of Nfix prevents fetal and maintains embryonic gene expression in the fetus. Therefore, Nfix acts as a transcriptional switch from embryonic to fetal myogenesis.


Asunto(s)
Desarrollo de Músculos , Músculo Esquelético/embriología , Factores de Transcripción NFI/metabolismo , Transcripción Genética , Animales , Feto/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Isoenzimas/metabolismo , Factores de Transcripción MEF2 , Ratones , Factores Reguladores Miogénicos/metabolismo , Factores de Transcripción NFATC/metabolismo , Factor de Transcripción PAX7/metabolismo , Fosfopiruvato Hidratasa , Proteína Quinasa C/metabolismo , Proteína Quinasa C-theta
10.
Dev Biol ; 490: 134-143, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35917935

RESUMEN

The vertebrate embryonic midline vasculature forms in close proximity to the developing skeletal muscle, which originates in the somites. Angioblasts migrate from bilateral positions along the ventral edge of the somites until they meet at the midline, where they sort and differentiate into the dorsal aorta and the cardinal vein. This migration occurs at the same time that myoblasts in the somites are beginning to differentiate into skeletal muscle, a process which requires the activity of the basic helix loop helix (bHLH) transcription factors Myod and Myf5. Here we examined vasculature formation in myod and myf5 mutant zebrafish. In the absence of skeletal myogenesis, angioblasts migrate normally to the midline but form only the cardinal vein and not the dorsal aorta. The phenotype is due to the failure to activate vascular endothelial growth factor ligand vegfaa expression in the somites, which in turn is required in the adjacent angioblasts for dorsal aorta specification. Myod and Myf5 cooperate with Hedgehog signaling to activate and later maintain vegfaa expression in the medial somites, which is required for angiogenic sprouting from the dorsal aorta. Our work reveals that the early embryonic skeletal musculature in teleosts evolved to organize the midline vasculature during development.


Asunto(s)
Proteína MioD , Factores Reguladores Miogénicos , Animales , Aorta/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas Musculares/genética , Músculo Esquelético , Proteína MioD/genética , Proteína MioD/metabolismo , Factor 5 Regulador Miogénico/genética , Factor 5 Regulador Miogénico/metabolismo , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
11.
FASEB J ; 35(4): e21346, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33715228

RESUMEN

Dynamin 2 (DNM2) is a ubiquitously expressed protein involved in many functions related to trafficking and remodeling of membranes and cytoskeleton dynamics. Mutations in the DNM2 gene cause the autosomal dominant centronuclear myopathy (AD-CNM), characterized mainly by muscle weakness and central nuclei. Several defects have been identified in the KI-Dnm2R465W/+ mouse model of the disease to explain the muscle phenotype, including reduction of the satellite cell pool in muscle, but the functional consequences of this depletion have not been characterized until now. Satellite cells (SC) are the main source for muscle growth and regeneration of mature tissue. Here, we investigated muscle regeneration in the KI-Dnm2R465W/+ mouse model for AD-CNM. We found a reduced number of Pax7-positive SCs, which were also less activated after induced muscle injury. The muscles of the KI-Dnm2R465W/+ mouse regenerated more slowly and less efficiently than wild-type ones, formed fewer new myofibers, and did not recover its normal mass 15 days after injury. Altogether, our data provide evidence that the muscle regeneration is impaired in the KI-Dnm2R465W/+ mouse and contribute with one more layer to the comprehension of the disease, by identifying a new pathomechanism linked to DNM2 mutations which may be involved in the muscle-specific impact occurring in AD-CNM.


Asunto(s)
Dinamina II/metabolismo , Músculo Esquelético/lesiones , Miopatías Estructurales Congénitas/genética , Células Satélite del Músculo Esquelético/fisiología , Animales , Dinamina II/genética , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Ratones , Mutación , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Regeneración
12.
J Muscle Res Cell Motil ; 43(1): 9-20, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35018575

RESUMEN

Skeletal muscle contractions are caused to release myokines by muscle fiber. This study investigated the myogenic regulatory factors, as MHC I, IIA, IIX, Myo-D, MRF4, Murf, Atrogin-1, Decorin, Myonection, and IL-15 mRNA expression in the response of eccentric vs concentric contraction. Eighteen healthy men were randomly divided into two eccentric and concentric groups, each of 9 persons. Isokinetic contraction protocols included maximal single-leg eccentric or concentric knee extension tasks at 60°/s with the dominant leg. Contractions consisted of a maximum of 12 sets of 10 reps, and the rest time between each set was 30 s. The baseline biopsy was performed 4 weeks before the study, and post-test biopsies were taken immediately after exercise protocols from the vastus lateralis muscle. The gene expression levels were evaluated using Real-Time PCR methods. The eccentric group showed a significantly lower RPE score than the concentric group (P ≤ 0.05). A significant difference in MyoD, MRF4, Myonection, and Decorin mRNA, were observed following eccentric or concentric contractions (P ≤ 0.05). The MHC I, MHC IIA, IL-15 mRNA has been changed significantly compared to the pre-exercise in the concentric group (P ≤ 0.05). While only MHC IIX and Atrogin-1 mRNA changed significantly in the eccentric group (P ≤ 0.05). Additionally, the results showed a significant difference in MyoD, MRF4, IL-15, and Decorin at the follow-up values between eccentric or concentric groups (P ≤ 0.05). Our findings highlight the growing importance of elucidating the different responses of muscle growth factors associated with a myogenic activity such as MHC IIA, Decorin, IL-15, Myonectin, Decorin, MuRF1, and MHC IIX mRNA in following various types of exercise.


Asunto(s)
Factores Reguladores Miogénicos , Músculo Cuádriceps , Decorina/genética , Decorina/metabolismo , Humanos , Interleucina-15/genética , Interleucina-15/metabolismo , Masculino , Contracción Muscular/fisiología , Músculo Esquelético/metabolismo , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Músculo Cuádriceps/metabolismo , ARN Mensajero/metabolismo
13.
Biomarkers ; 27(8): 753-763, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35946424

RESUMEN

BACKGROUND: The present study aimed to analyse the role of myokines and the regeneration capacity of skeletal muscle during chronic hypobaric hypoxia (CHH). METHODS: Male Sprague-Dawley rats were exposed to hypobaric hypoxia (HH) for 1d, 3d and 7d. RESULTS: Exposure to HH enhanced the levels of decorin, irisin, IL-6 and IL-15 till 3 days of hypoxia and on 7 day of exposure, no significant changes were observed in relation to control. A significant upregulation in myostatin, activated protein kinase, SMAD3, SMAD4, FOXO-1, MURF-1 expression was observed with prolonged HH exposure as compared to normoxic control. Further, myogenesis-related markers, PAX-7, Cyclin D1 and myogenin were downregulated during CHH exposure in comparison to control. Energy metabolism regulators such as Sirtuin 1, proliferator-activated receptor gamma coactivator-1α and GLUT-4, were also increased on 1-d HH exposure that showed a declining trend on CHH exposure. CONCLUSIONS: These results indicated the impairment in the levels of myokines and myogenesis during prolonged hypoxia. CHH exposure enhanced the levels of myostatin and reduced the regeneration or repair capacity of the skeletal muscles. Myokine levels could be a predictive biomarker for evaluating skeletal muscle performance and loss at high altitudes.


Asunto(s)
Factores Reguladores Miogénicos , Miostatina , Ratas , Animales , Masculino , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Ratas Sprague-Dawley , Hipoxia , Músculo Esquelético
14.
Anim Biotechnol ; 33(6): 1095-1108, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33491546

RESUMEN

The detection of activin receptor typeIIB (ACTRIIB) protein, a prominent negative muscle growth regulator has paramount value in augmenting growth traits through molecular breeding schemes in chicken. The study was formulated to establish primary chicken embryo myoblast culture (CEM) using 9th and 18th day chick embryos and to develop antibodies for immunodetection of ACTRIIB protein. The physicochemical and structural attributes of the ACTRIIB sequence were evaluated to identify substantial antigenic regions. The ACTRIIB sequence was transfected into CEM and expressed protein was injected subcutaneously into rats to produce hyperimmune serum. The average propensity of protein sequence for beta turns, surface accessibility, chain flexibility, antigenicity, hydrophilicity and linear epitopes was 0.978, 1.000, 0.991, 1.038, 1.258 and 0.512, respectively. The 9th day CEM exhibited confluency (80-90%) earlier than the 18th day. The expression of myogenic regulatory factors in 9th day myoblasts was higher than the 18th day by 7.28, 5.16, 6.28 and 6.93 folds for MYF5, MRF4, MYOG and MYOD, respectively. The ACTRIIB mRNA was downregulated by 2.54 folds on the 9th day compared to the 18th day myoblasts and protein varied significantly between 9th and 18th day myoblasts. The CEM culture can be harnessed unequivocally to investigate molecular mechanisms underlying muscle growth besides raising antibodies.


Asunto(s)
Pollos , Mioblastos , Embrión de Pollo , Ratas , Animales , Pollos/genética , Epítopos/metabolismo , Mioblastos/metabolismo , Factores Reguladores Miogénicos/metabolismo , Técnicas de Cultivo de Célula
15.
Int J Mol Sci ; 23(13)2022 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-35805999

RESUMEN

Epinephelus coioides is a fish species with high economic value due to its delicious meat, high protein content, and rich fatty acid nutrition. It has become a high-economic fish in southern parts of China and some other Southeast Asian countries. In this study, the myostatin nucleic acid vaccine was constructed and used to immunize E. coioides. The results from body length and weight measurements indicated the myostatin nucleic acid vaccine promoted E. coioides growth performance by increasing muscle fiber size. The results from RT-qPCR analysis showed that myostatin nucleic acid vaccine upregulated the expression of myod, myog and p21 mRNA, downregulated the expression of smad3 and mrf4 mRNA. This preliminary study is the first report that explored the role of myostatin in E. coioides and showed positive effects of autologous nucleic acid vaccine on the muscle growth of E. coioides. Further experiments with increased numbers of animals and different doses are needed for its application to E. coiodes aquaculture production.


Asunto(s)
Fibras Musculares Esqueléticas , Miostatina , Perciformes , Animales , Peso Corporal , Peces , Regulación de la Expresión Génica , Fibras Musculares Esqueléticas/fisiología , Proteína MioD/genética , Proteína MioD/metabolismo , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Miogenina/genética , Miogenina/metabolismo , Miostatina/genética , Miostatina/inmunología , Vacunación Basada en Ácidos Nucleicos/administración & dosificación , Vacunación Basada en Ácidos Nucleicos/inmunología , Perciformes/crecimiento & desarrollo , Perciformes/fisiología , Proteína smad3/genética , Proteína smad3/metabolismo , Vacunación , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo
16.
Dev Biol ; 468(1-2): 133-145, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32768399

RESUMEN

The importance of skeletal muscle for rib development and patterning in the mouse embryo has not been resolved, largely because different experimental approaches have yielded disparate results. In this study, we utilize both gene knockouts and muscle cell ablation approaches to re-visit the extent to which rib growth and patterning are dependent on developing musculature. Consistent with previous studies, we show that rib formation is highly dependent on the MYOD family of myogenic regulatory factors (MRFs), and demonstrate that the extent of rib formation is gene-, allele-, and dosage-dependent. In the absence of Myf5 and MyoD, one allele of Mrf4 is sufficient for extensive rib growth, although patterning is abnormal. Under conditions of limiting MRF dosage, MyoD is identified as a positive regulator of rib patterning, presumably due to improved intercostal muscle development. In contrast to previous muscle ablation studies, we show that diphtheria toxin subunit A (DTA)-mediated ablation of muscle progenitors or differentiated muscle, using MyoDiCre or HSA-Cre drivers, respectively, profoundly disrupts rib development. Further, a comparison of three independently derived Rosa26-based DTA knockin alleles demonstrates that the degree of rib perturbations in MyoDiCre/+/DTA embryos is markedly dependent on the DTA allele used, and may in part explain discrepancies with previous findings. The results support the conclusion that the extent and quality of rib formation is largely dependent on the dosage of Myf5 and Mrf4, and that both early myotome-sclerotome interactions, as well as later muscle-rib interactions, are important for proper rib growth and patterning.


Asunto(s)
Tipificación del Cuerpo , Músculo Esquelético/embriología , Costillas/embriología , Alelos , Animales , Hormona Liberadora de Gonadotropina/análogos & derivados , Ratones Transgénicos , Proteína MioD/genética , Proteína MioD/metabolismo , Factor 5 Regulador Miogénico/genética , Factor 5 Regulador Miogénico/metabolismo , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo
17.
Nat Immunol ; 10(3): 289-96, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19169261

RESUMEN

Despite advances in the identification of lymphoid-restricted progenitor cells, the transcription factors essential for their generation remain to be identified. Here we describe an unexpected function for the myeloid oncogene product Mef2c in lymphoid development. Mef2c deficiency was associated with profound defects in the production of B cells, T cells, natural killer cells and common lymphoid progenitor cells and an enhanced myeloid output. In multipotent progenitors, Mef2c was required for the proper expression of several key lymphoid regulators and restriction of the myeloid fate. Our studies also show that Mef2c was a critical transcriptional target of the transcription factor PU.1 during lymphopoiesis. Thus, Mef2c is a crucial component of the transcriptional network that regulates cell fate 'choice' in multipotent progenitors.


Asunto(s)
Células Progenitoras Linfoides/metabolismo , Linfopoyesis , Factores Reguladores Miogénicos/metabolismo , Animales , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Factores de Transcripción MEF2 , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores Reguladores Miogénicos/genética , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo
18.
Clin Exp Dermatol ; 46(7): 1255-1261, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33899950

RESUMEN

BACKGROUND: Keloid lesions are characterized by mesenchymal cell proliferation and excessive extracellular matrix deposition. Previous microarray analyses have been performed to investigate the mechanism of keloid development. However, the molecular pathology that contributes to keloid development remains obscure. AIM: To explore the underlying essential molecules of keloids using microarrays. METHODS: We performed microarray analyses of keloid and nonlesional skin tissues both in vivo and in vitro. Gene expression levels were compared between tissues and cells. Quantitative reverse transcription (qRT)-PCR and immunohistochemical staining were used to determine the expression levels of molecules of interest in keloid tissues. RESULTS: Several common molecules were upregulated in both keloid tissues and keloid-lesional fibroblasts. PTPRD and NTM were upregulated both in vivo and in vitro. The genes MDFI and ITGA4 were located at the centre of the gene coexpression network analysis using keloid tissues. qRT-PCR revealed significant expression levels of PTPRD and MDFI in keloid tissues. Immunopathological staining revealed that MDFI-positive cells, which have fibroblast characteristics, were located in the keloid-associated lymphoid tissue (KALT) portion of the keloid tissue. CONCLUSION: Our gene expression profiles of keloids could distinguish the difference between lesional tissue and cultured lesional fibroblasts, and MDFI was found to be commonly expressed in both tissues and cells. Thus, MDFI-positive cells, which were located in the KALT, may play an important role in keloid pathogenesis and thus might be useful for in vitro keloid studies.


Asunto(s)
Perfilación de la Expresión Génica , Expresión Génica , Queloide/genética , Factores Reguladores Miogénicos/genética , Diagnóstico Diferencial , Fibroblastos/metabolismo , Humanos , Inmunohistoquímica , Queloide/metabolismo , Análisis por Micromatrices , Factores Reguladores Miogénicos/metabolismo , ARN/análisis , ARN Mensajero/metabolismo , Regulación hacia Arriba
19.
Genes Dev ; 27(11): 1247-59, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23723416

RESUMEN

Alternate splicing contributes extensively to cellular complexity by generating protein isoforms with divergent functions. However, the role of alternate isoforms in development remains poorly understood. Mef2 transcription factors are essential transducers of cell signaling that modulate differentiation of many cell types. Among Mef2 family members, Mef2D is unique, as it undergoes tissue-specific splicing to generate a muscle-specific isoform. Since the ubiquitously expressed (Mef2Dα1) and muscle-specific (Mef2Dα2) isoforms of Mef2D are both expressed in muscle, we examined the relative contribution of each Mef2D isoform to differentiation. Using both in vitro and in vivo models, we demonstrate that Mef2D isoforms act antagonistically to modulate differentiation. While chromatin immunoprecipitation (ChIP) sequencing analysis shows that the Mef2D isoforms bind an overlapping set of genes, only Mef2Dα2 activates late muscle transcription. Mechanistically, the differential ability of Mef2D isoforms to activate transcription depends on their susceptibility to phosphorylation by protein kinase A (PKA). Phosphorylation of Mef2Dα1 by PKA provokes its association with corepressors. Conversely, exon switching allows Mef2Dα2 to escape this inhibitory phosphorylation, permitting recruitment of Ash2L for transactivation of muscle genes. Thus, our results reveal a novel mechanism in which a tissue-specific alternate splicing event has evolved that permits a ubiquitously expressed transcription factor to escape inhibitory signaling for temporal regulation of gene expression.


Asunto(s)
Empalme Alternativo , Diferenciación Celular/genética , Músculos/citología , Músculos/metabolismo , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Animales , Inmunoprecipitación de Cromatina , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de Unión al ADN/metabolismo , Exones/genética , Regulación de la Expresión Génica/genética , Genoma/genética , Factores de Transcripción MEF2 , Ratones , Músculos/enzimología , Mutación/genética , Factores Reguladores Miogénicos/química , Proteínas Nucleares/metabolismo , Especificidad de Órganos/genética , Fosforilación/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Transcripción Genética/genética
20.
Int J Mol Sci ; 22(7)2021 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-33918414

RESUMEN

The process of myogenesis gradually deteriorates as the skeletal muscle ages, contributing to muscle mass loss. The aim of this study is to investigate the effect of senescence/aging on skeletal myogenesis, in vitro. A model of multiple cell divisions of C2C12 myoblasts was used to replicate cell senescence. Control and aged myoblasts were investigated during myogenesis, i.e., at days 0, 2, and 6of differentiation. SA-ß-gal activity and comet assay were used as markers of aging and DNA damage. Flow cytometry was performed to characterize potential differences in cell cycle between control and aged cells. Alterations in the mRNA and/or protein expression of myogenic regulatory factors (MRFs), IGF-1 isoforms, apoptotic, atrophy, inflammatory, metabolic and aging-related factors were evaluated. Compared with the control cells, aged myoblasts exhibited G0/G1 cell cycle arrest, DNA damage, increased SA-ß-gal activity, and increased expression of aging-related factors p16 and p21 during differentiation. Moreover, aged myoblasts showed a reduction in the expression of MRFs and metabolic/anabolic factors, along with an increased expression of apoptotic, atrophy and inflammatory factors. A diminished differentiation capacity characterized the aged myoblasts which, in combination with the induction of apoptotic and atrophy factors, indicated a disrupted myogenic lineage in the senescent muscle cells.


Asunto(s)
Senescencia Celular , Desarrollo de Músculos , Animales , Línea Celular , Ratones , Mioblastos/metabolismo , Factores Reguladores Miogénicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA