Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2023 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-37293078

RESUMEN

Alanyl-transfer RNA synthetase 2 (AARS2) is a nuclear encoded mitochondrial tRNA synthetase that is responsible for charging of tRNA-Ala with alanine during mitochondrial translation. Homozygous or compound heterozygous mutations in the Aars2 gene, including those affecting its splicing, are linked to infantile cardiomyopathy in humans. However, how Aars2 regulates heart development, and the underlying molecular mechanism of heart disease remains unknown. Here, we found that poly(rC) binding protein 1 (PCBP1) interacts with the Aars2 transcript to mediate its alternative splicing and is critical for the expression and function of Aars2. Cardiomyocyte-specific deletion of Pcbp1 in mice resulted in defects in heart development that are reminiscent of human congenital cardiac defects, including noncompaction cardiomyopathy and a disruption of the cardiomyocyte maturation trajectory. Loss of Pcbp1 led to an aberrant alternative splicing and a premature termination of Aars2 in cardiomyocytes. Additionally, Aars2 mutant mice with exon-16 skipping recapitulated heart developmental defects observed in Pcbp1 mutant mice. Mechanistically, we found dysregulated gene and protein expression of the oxidative phosphorylation pathway in both Pcbp1 and Aars2 mutant hearts; these date provide further evidence that the infantile hypertrophic cardiomyopathy associated with the disorder oxidative phosphorylation defect type 8 (COXPD8) is mediated by Aars2. Our study therefore identifies Pcbp1 and Aars2 as critical regulators of heart development and provides important molecular insights into the role of disruptions in metabolism on congenital heart defects.

2.
J Biol Chem ; 292(23): 9540-9550, 2017 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-28381556

RESUMEN

Regulation of gene expression during muscle development and disease remains incompletely understood. microRNAs are a class of small non-coding RNAs that regulate gene expression and function post-transcriptionally. The poly(C)-binding protein1 (Pcbp1, hnRNP-E1, or αCP-1) is an RNA-binding protein that has been reported to bind the 3'-UTRs of target genes to regulate mRNA stability and protein translation. However, Pcbp1's biological function and the general mechanism of action remain largely undetermined. Here, we report that Pcbp1 is a component of the miRNA-processing pathway that regulates miRNA biogenesis. siRNA-based inhibition of Pcbp1 in mouse skeletal muscle myoblasts led to dysregulated cellular proliferation and differentiation. We also found that Pcbp1 null mutant mice exhibit early embryonic lethality, indicating that Pcbp1 is indispensable for embryonic development. Interestingly, hypomorphic Pcbp1 mutant mice displayed defects in muscle growth due to defects in the proliferation and differentiation of myoblasts and muscle satellite cells, in addition to a slow to fast myofibril switch. Moreover, Pcbp1 modulated the processing of muscle-enriched miR-1, miR-133, and miR-206 by physically interacting with argonaute 2 (AGO2) and other miRNA pathway components. Our study, therefore, uncovers the important function of Pcbp1 in skeletal muscle and the microRNA pathway, signifying its potential as a therapeutic target for muscle disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , MicroARNs/metabolismo , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/metabolismo , Procesamiento Postranscripcional del ARN/fisiología , Estabilidad del ARN/fisiología , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Proteínas Portadoras/genética , Línea Celular , Proteínas de Unión al ADN , Ratones , MicroARNs/genética , Proteínas de Unión al ARN , Transducción de Señal/fisiología
3.
Genesis ; 52(10): 879-87, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25174608

RESUMEN

The molecular events that control cell fate determination in cardiac and smooth muscle lineages remain elusive. Myocardin is an important transcription cofactor that regulates cell proliferation, differentiation, and development of the cardiovascular system. Here, we describe the construction and analysis of a dual Cre and enhanced green fluorescent protein (EGFP) knock-in mouse line in the Myocardin locus (Myocd(KI)). We report that the Myocd(KI) allele expresses the Cre enzyme and the EGFP in a manner that recapitulates endogenous Myocardin expression patterns. We show that Myocardin expression marks the earliest cardiac and smooth muscle lineages. Furthermore, this genetic model allows for the identification of a cardiac cell population, which maintains both Myocardin and Isl-1 expression, in E7.75-E8.0 embryos, highlighting the contribution and merging of the first and second heart fields during cardiogenesis. Therefore, the Myocd(KI) allele is a unique tool for studying cardiovascular development and lineage-specific gene manipulation.


Asunto(s)
Linaje de la Célula , Integrasas/genética , Miocitos Cardíacos/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Animales , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Técnicas de Sustitución del Gen , Sitios Genéticos , Proteínas con Homeodominio LIM/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos , Factores de Transcripción/metabolismo
4.
J Am Heart Assoc ; 3(3): e000796, 2014 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-24847033

RESUMEN

BACKGROUND: Inactivation of Shox2, a member of the short-stature homeobox gene family, leads to defective development of multiple organs and embryonic lethality as a result of cardiovascular defects, including bradycardia and severe hypoplastic sinoatrial node (SAN) and sinus valves, in mice. It has been demonstrated that Shox2 regulates a genetic network through the repression of Nkx2.5 to maintain the fate of the SAN cells. However, the functional mechanism of Shox2 protein as a transcriptional repressor on Nkx2.5 expression remains completely unknown. METHODS AND RESULTS: A specific interaction between the B56δ regulatory subunit of PP2A and Shox2a, the isoform that is expressed in the developing heart, was demonstrated by yeast 2-hybrid screen and coimmunoprecipitation. Western blotting and immunohistochemical assays further confirmed the presence of phosphorylated Shox2a (p-Shox2a) in cell culture as well as in the developing mouse and human SAN. Site-directed mutagenesis and in vitro kinase assays identified Ser92 and Ser110 as true phosphorylation sites and substrates of extracellular signal-regulated kinase 1 and 2. Despite that Shox2a and its phosphorylation mutants possessed similar transcriptional repressive activities in cell cultures when fused with Gal4 protein, the mutant forms exhibited a compromised repressive effect on the activity of the mouse Nkx2.5 promoter in cell cultures, indicating that phosphorylation is required for Shox2a to repress Nkx2.5 expression specifically. Transgenic expression of Shox2a, but not Shox2a-S92AS110A, mutant in the developing heart resulted in down-regulation of Nkx2.5 in wild-type mice and rescued the SAN defects in the Shox2 mutant background. Last, we demonstrated that elimination of both phosphorylation sites on Shox2a did not alter its nuclear location and dimerization, but depleted its capability to bind to the consensus sequences within the Nkx2.5 promoter region. CONCLUSIONS: Our studies reveal that phosphorylation is essential for Shox2a to repress Nkx2.5 expression during SAN development and differentiation.


Asunto(s)
Proteínas de Homeodominio/fisiología , Nodo Sinoatrial/crecimiento & desarrollo , Animales , Células Cultivadas , Ensayo de Cambio de Movilidad Electroforética , Regulación del Desarrollo de la Expresión Génica/fisiología , Frecuencia Cardíaca/fisiología , Proteína Homeótica Nkx-2.5 , Humanos , Ratones , Ratones Transgénicos , Mutagénesis Sitio-Dirigida/métodos , Fosforilación/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Nodo Sinoatrial/embriología , Factores de Transcripción/fisiología , Técnicas del Sistema de Dos Híbridos
5.
Curr Top Dev Biol ; 100: 279-317, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22449848

RESUMEN

MicroRNAs (miRNAs) are a class of small noncoding RNAs of ~22nt in length which are involved in the regulation of gene expression at the posttranscriptional level by degrading their target mRNAs and/or inhibiting their translation. Expressed ubiquitously or in a tissue-specific manner, miRNAs are involved in the regulation of many biological processes such as cell proliferation, differentiation, apoptosis, and the maintenance of normal cellular physiology. Many miRNAs are expressed in embryonic, postnatal, and adult hearts. Aberrant expression or genetic deletion of miRNAs is associated with abnormal cardiac cell differentiation, disruption of heart development, and cardiac dysfunction. This chapter will summarize the history, biogenesis, and processing of miRNAs as well as their function in heart development, remodeling, and disease.


Asunto(s)
Corazón/embriología , Corazón/crecimiento & desarrollo , MicroARNs/genética , Animales , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Cardiopatías/genética , Cardiopatías/patología , Humanos , Miocardio/citología , Miocardio/metabolismo
6.
Pediatr Cardiol ; 33(6): 882-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22307400

RESUMEN

Embryonic development is a tightly regulated process, and many families of genes functions to provide a regulatory genetic network to achieve such a program. The homeobox genes are an extensive family that encodes transcription factors with a characteristic 60-amino acid homeodomain. Mutations in these genes or in the encoded proteins might result in structural malformations, physiological defects, and even embryonic death. Mutations in the short-stature homeobox gene (SHOX) is associated with idiopathic short stature in humans, as observed in patients with Turner syndrome and/or Leri-Weill dyschondrosteosis. A closely related human homolog, SHOX2, has not been linked to any syndrome or defect so far. In mice, a SHOX ortholog gene is not present in the genome; however, a true SHOX2 ortholog has been identified. Analyses of Shox2 knockout mouse models have showed crucial functions during embryonic development, including limb skeletogenesis, palatogenesis, temporomandibular joint formation, and cardiovascular development. During embryonic cardiac development, Shox2 is restrictedly expressed in the sinus venosus region, including the sinoatrial node (SAN) and the sinus valves. Shox2 null mutant is embryonically lethal due to cardiovascular defects, including a severely hypoplastic SAN and sinus valves attributed to a significantly decreased level of cell proliferation in addition to an abnormal low heartbeat rate (bradycardia). In addition, it has been demonstrated that Shox2 regulates a genetic network through the repression of Nkx2.5 to maintain the SAN fate and thus plays essential roles in its proper formation and differentiation.


Asunto(s)
Corazón/embriología , Proteínas de Homeodominio/metabolismo , Nodo Sinoatrial/embriología , Animales , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox , Proteínas de Homeodominio/genética , Humanos , Ratones , Proteína de la Caja Homeótica de Baja Estatura , Factores de Transcripción
7.
Dev Biol ; 356(2): 359-69, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21640717

RESUMEN

The sinoatrial node (SAN), functionally known as the pacemaker, regulates the cardiac rhythm or heartbeat. Several genes are expressed in the developing SAN and form a genetic network regulating the fate of the SAN cells. The short stature homeobox gene Shox2 is an important player in the SAN genetic network by regulating the expression of different cardiac conduction molecular markers including the early cardiac differentiation marker Nkx2.5. Here we report that the expression patterns of Shox2 and Nkx2.5 are mutually exclusive from the earliest stages of the venous pole and the SAN formation. We show that tissue specific ectopic expression of Shox2 in the developing mouse heart downregulates the expression of Nkx2.5 and causes cardiac malformations; however, it is not sufficient to induce a SAN cell fate switch in the working myocardium. On the other hand, tissue specific overexpression of Nkx2.5 in the heart leads to severe hypoplasia of the SAN and the venous valves, dis-regulation of the SAN genetic network, and change of the SAN cell fate into working myocardium, and causes embryonic lethality, recapitulating the phenotypes including bradycardia observed in Shox2(-/-) mutants. These results indicate that Nkx2.5 activity is detrimental to the normal formation of the SAN. Taken together, our results demonstrate that Shox2 downregulation of Nkx2.5 is essential for the proper development of the SAN and that Shox2 functions to shield the SAN from becoming working myocardium by acting upstream of Nkx2.5.


Asunto(s)
Proteínas de Homeodominio/fisiología , Nodo Sinoatrial/embriología , Factores de Transcripción/fisiología , Animales , Redes Reguladoras de Genes , Cardiopatías Congénitas/etiología , Proteína Homeótica Nkx-2.5 , Ratones , Especificidad de Órganos
8.
J Biol Chem ; 286(19): 17029-38, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21454626

RESUMEN

The homeodomain transcription factor Shox2 plays a crucial regulatory role in the development of sinoatrial node (SAN) by repressing the expression of Nkx2.5, as demonstrated by failed differentiation of SAN in Shox2 null mice. The SHOX (short stature homeobox) gene family consists of two closely related members, SHOX and SHOX2 in humans, but a SHOX ortholog does not exist in the mouse genome. These two genes exhibit overlapping and distinct expression patterns in many developing organs but whether they share functional redundancy is not known. In this study, we set to investigate possible functional redundancy between SHOX and SHOX2 in vitro and in vivo. We first showed that human SHOX and SHOX2 and mouse Shox2 possess similar transcriptional repressive activities in cell cultures, particularly the repressive effects on the Nkx2.5 promoter activity. We further created an SHOX/Shox2 knock-in mouse line (replacement of Shox2 with SHOX, referred as Shox2(KI/KI)). Mice carrying the hypomorphic Shox2(KI+Neo/KI+Neo) allele exhibit bradycardia and arrhythmia and die a few days after birth. However, mice carrying the Shox2(KI/KI) allele grow to adulthood. Physiological, histological, and molecular analyses demonstrate a fully developed SAN and normal pacemaking function in Shox2(KI/KI) mice. Our results demonstrate a functional redundancy between human SHOX and mouse Shox2 in the regulation of SAN formation and pacemaking function in addition to several other organs. The SHOX/Shox2 dose appears to be critical for normal pacemaking function.


Asunto(s)
Regulación de la Expresión Génica , Corazón/fisiología , Proteínas de Homeodominio/metabolismo , Nodo Sinoatrial/metabolismo , Alelos , Animales , Arritmias Cardíacas/metabolismo , Electrocardiografía/métodos , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Genéticos , Proteína de la Caja Homeótica de Baja Estatura , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Cell Tissue Res ; 342(3): 401-10, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21079999

RESUMEN

The skin is the outer layer of protection against the environment. The development and formation of the skin is regulated by several genetic cascades including the bone morphogenetic protein (BMP) signaling pathway, which has been suggested to play an important role during embryonic organ development. Several skin defects and diseases are caused by genetic mutations or disorders. Ichthyosis is a common genetic skin disorder characterized by dry scaly skin. Loss-of-function mutations in the filaggrin (FLG) gene have been identified as the cause of the ichthyosis vulgaris (IV) phenotype; however, the direct regulation of filaggrin expression in vivo is unknown. We present evidence that BMP signaling regulates filaggrin expression in the epidermis. Mice expressing a constitutively active form of BMP-receptor-IB in the developing epidermis exhibit a phenotype resembling IV in humans, including dry flaky skin, compact hyperkeratosis, and an attenuated granular layer associated with a significantly downregulated expression of filaggrin. Regulation of filaggrin expression by BMP signaling has been further confirmed by the application of exogenous BMP2 in skin explants and by a transgenic model overexpressing Noggin in the epidermis. Our results demonstrate that aberrant BMP signaling in the epidermis causes overproliferation and hyperkeratinization, leading to an IV-like skin disease.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Epidermis/metabolismo , Ictiosis Vulgar , Animales , Western Blotting , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Proteínas Portadoras/genética , Diferenciación Celular , Proliferación Celular , Epidermis/embriología , Epidermis/patología , Proteínas Filagrina , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Genotipo , Ictiosis Vulgar/genética , Ictiosis Vulgar/metabolismo , Ictiosis Vulgar/patología , Proteínas de Filamentos Intermediarios/genética , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones Transgénicos , Fenotipo , Reacción en Cadena de la Polimerasa , Transducción de Señal
10.
Dev Biol ; 327(2): 376-85, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19166829

RESUMEN

The pacemaker is composed of specialized cardiomyocytes located within the sinoatrial node (SAN), and is responsible for originating and regulating the heart beat. Recent advances towards understanding the SAN development have been made on the genetic control and gene interaction within this structure. Here we report that the Shox2 homeodomain transcription factor is restrictedly expressed in the sinus venosus region including the SAN and the sinus valves during embryonic heart development. Shox2 null mutation results in embryonic lethality due to cardiovascular defects, including an abnormal low heart beat rate (bradycardia) and severely hypoplastic SAN and sinus valves attributed to a significantly decreased level of cell proliferation. Genetically, the lack of Tbx3 and Hcn4 expression, along with ectopic activation of Nppa, Cx40, and Nkx2-5 in the Shox2(-/-) SAN region, indicates a failure in SAN differentiation. Furthermore, Shox2 overexpression in Xenopus embryos results in extensive repression of Nkx2-5 in the developing heart, leading to a reduced cardiac field and aberrant heart formation. Reporter gene expression assays provide additional evidence for the repression of Nkx2-5 promoter activity by Shox2. Taken together our results demonstrate that Shox2 plays an essential role in the SAN and pacemaker development by controlling a genetic cascade through the repression of Nkx2-5.


Asunto(s)
Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica , Corazón , Proteínas de Homeodominio/metabolismo , Nodo Sinoatrial , Factores de Transcripción/metabolismo , Animales , Femenino , Corazón/anatomía & histología , Corazón/embriología , Frecuencia Cardíaca , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nodo Sinoatrial/citología , Nodo Sinoatrial/embriología , Factores de Transcripción/genética , Xenopus laevis/anatomía & histología , Xenopus laevis/embriología
11.
Dev Dyn ; 237(12): 3613-23, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18985739

RESUMEN

Left-sided expression of Nodal in the lateral plate mesoderm (LPM) during early embryogenesis is a crucial step in establishing the left-right (L-R) axis in vertebrates. In the chick, it was suggested that chick Cerberus (cCer), a Cerberus/Dan family member, induces Nodal expression by antagonizing bone morphogenetic protein (BMP) activity in the left LPM. In contrast, it has also been shown that BMPs positively regulate Nodal expression in the left LPM in the chick embryo. Thus, it is still unclear how the bilaterally expressed BMPs induce Nodal expression only in the left LPM. In this study, we demonstrate that BMP signaling is necessary and sufficient for the induction of Nodal expression in the chick LPM where the type I BMP receptor-IB (BMPR-IB) likely mediates this induction. Tissue grafting experiments indicate the existence of a Nodal inductive factor in the left LPM rather than the presence of a Nodal inhibitory factor in the right LPM. We demonstrate that cCer functions as a BMP agonist instead of antagonist, being able to enhance BMP signaling in cell culture. This conclusion is further supported by the immunoprecipitation assays that provide convincing biochemical evidence for a direct interaction between cCer and BMP receptor. Because cCer is expressed restrictedly in the left LPM, BMPs and cCer appear to act synergistically to activate Nodal expression in the left LPM in the chick.


Asunto(s)
Proteínas Aviares/metabolismo , Tipificación del Cuerpo , Proteínas Morfogenéticas Óseas/agonistas , Proteínas Morfogenéticas Óseas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Nodal/metabolismo , Animales , Proteínas Aviares/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Diferenciación Celular , Línea Celular , Embrión de Pollo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...