Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Cell ; 185(5): 794-814.e30, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35182466

RESUMEN

Congenital heart disease (CHD) is present in 1% of live births, yet identification of causal mutations remains challenging. We hypothesized that genetic determinants for CHDs may lie in the protein interactomes of transcription factors whose mutations cause CHDs. Defining the interactomes of two transcription factors haplo-insufficient in CHD, GATA4 and TBX5, within human cardiac progenitors, and integrating the results with nearly 9,000 exomes from proband-parent trios revealed an enrichment of de novo missense variants associated with CHD within the interactomes. Scoring variants of interactome members based on residue, gene, and proband features identified likely CHD-causing genes, including the epigenetic reader GLYR1. GLYR1 and GATA4 widely co-occupied and co-activated cardiac developmental genes, and the identified GLYR1 missense variant disrupted interaction with GATA4, impairing in vitro and in vivo function in mice. This integrative proteomic and genetic approach provides a framework for prioritizing and interrogating genetic variants in heart disease.


Asunto(s)
Factor de Transcripción GATA4/metabolismo , Cardiopatías Congénitas , Proteínas Nucleares/metabolismo , Oxidorreductasas/metabolismo , Factores de Transcripción , Animales , Cardiopatías Congénitas/genética , Ratones , Mutación , Proteómica , Proteínas de Dominio T Box/genética , Factores de Transcripción/genética
2.
Development ; 150(9)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36994838

RESUMEN

Transcriptional networks governing cardiac precursor cell (CPC) specification are incompletely understood owing, in part, to limitations in distinguishing CPCs from non-cardiac mesoderm in early gastrulation. We leveraged detection of early cardiac lineage transgenes within a granular single-cell transcriptomic time course of mouse embryos to identify emerging CPCs and describe their transcriptional profiles. Mesp1, a transiently expressed mesodermal transcription factor, is canonically described as an early regulator of cardiac specification. However, we observed perdurance of CPC transgene-expressing cells in Mesp1 mutants, albeit mislocalized, prompting us to investigate the scope of the role of Mesp1 in CPC emergence and differentiation. Mesp1 mutant CPCs failed to robustly activate markers of cardiomyocyte maturity and crucial cardiac transcription factors, yet they exhibited transcriptional profiles resembling cardiac mesoderm progressing towards cardiomyocyte fates. Single-cell chromatin accessibility analysis defined a Mesp1-dependent developmental breakpoint in cardiac lineage progression at a shift from mesendoderm transcriptional networks to those necessary for cardiac patterning and morphogenesis. These results reveal Mesp1-independent aspects of early CPC specification and underscore a Mesp1-dependent regulatory landscape required for progression through cardiogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Epigenómica , Miocitos Cardíacos , Animales , Ratones , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica , Mesodermo/metabolismo , Miocitos Cardíacos/metabolismo , Factores de Transcripción/metabolismo
3.
Nature ; 571(7763): 107-111, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31217582

RESUMEN

Large-scale genome sequencing is poised to provide a substantial increase in the rate of discovery of disease-associated mutations, but the functional interpretation of such mutations remains challenging. Here we show that deletions of a sequence on human chromosome 16 that we term the intestine-critical region (ICR) cause intractable congenital diarrhoea in infants1,2. Reporter assays in transgenic mice show that the ICR contains a regulatory sequence that activates transcription during the development of the gastrointestinal system. Targeted deletion of the ICR in mice caused symptoms that recapitulated the human condition. Transcriptome analysis revealed that an unannotated open reading frame (Percc1) flanks the regulatory sequence, and the expression of this gene was lost in the developing gut of mice that lacked the ICR. Percc1-knockout mice displayed phenotypes similar to those observed upon ICR deletion in mice and patients, whereas an ICR-driven Percc1 transgene was sufficient to rescue the phenotypes found in mice that lacked the ICR. Together, our results identify a gene that is critical for intestinal function and underscore the need for targeted in vivo studies to interpret the growing number of clinical genetic findings that do not affect known protein-coding genes.


Asunto(s)
Diarrea/congénito , Diarrea/genética , Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica , Genes , Intestinos/fisiología , Eliminación de Secuencia/genética , Animales , Cromosomas Humanos Par 16/genética , Modelos Animales de Enfermedad , Femenino , Genes Reporteros , Sitios Genéticos/genética , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Linaje , Fenotipo , Activación Transcripcional , Transcriptoma/genética , Transgenes/genética
4.
Circulation ; 146(10): 770-787, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-35938400

RESUMEN

BACKGROUND: GATA4 (GATA-binding protein 4), a zinc finger-containing, DNA-binding transcription factor, is essential for normal cardiac development and homeostasis in mice and humans, and mutations in this gene have been reported in human heart defects. Defects in alternative splicing are associated with many heart diseases, yet relatively little is known about how cell type- or cell state-specific alternative splicing is achieved in the heart. Here, we show that GATA4 regulates cell type-specific splicing through direct interaction with RNA and the spliceosome in human induced pluripotent stem cell-derived cardiac progenitors. METHODS: We leveraged a combination of unbiased approaches including affinity purification of GATA4 and mass spectrometry, enhanced cross-linking with immunoprecipitation, electrophoretic mobility shift assays, in vitro splicing assays, and unbiased transcriptomic analysis to uncover GATA4's novel function as a splicing regulator in human induced pluripotent stem cell-derived cardiac progenitors. RESULTS: We found that GATA4 interacts with many members of the spliceosome complex in human induced pluripotent stem cell-derived cardiac progenitors. Enhanced cross-linking with immunoprecipitation demonstrated that GATA4 also directly binds to a large number of mRNAs through defined RNA motifs in a sequence-specific manner. In vitro splicing assays indicated that GATA4 regulates alternative splicing through direct RNA binding, resulting in functionally distinct protein products. Correspondingly, knockdown of GATA4 in human induced pluripotent stem cell-derived cardiac progenitors resulted in differential alternative splicing of genes involved in cytoskeleton organization and calcium ion import, with functional consequences associated with the protein isoforms. CONCLUSIONS: This study shows that in addition to its well described transcriptional function, GATA4 interacts with members of the spliceosome complex and regulates cell type-specific alternative splicing via sequence-specific interactions with RNA. Several genes that have splicing regulated by GATA4 have functional consequences and many are associated with dilated cardiomyopathy, suggesting a novel role for GATA4 in achieving the necessary cardiac proteome in normal and stress-responsive conditions.


Asunto(s)
Factor de Transcripción GATA4 , Células Madre Pluripotentes Inducidas , Empalme Alternativo , Animales , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Corazón , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , ARN/genética , ARN/metabolismo
5.
Cell ; 135(6): 1053-64, 2008 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-19070576

RESUMEN

Vascular development begins when mesodermal cells differentiate into endothelial cells, which then form primitive vessels. It has been hypothesized that endothelial-specific gene expression may be regulated combinatorially, but the transcriptional mechanisms governing specificity in vascular gene expression remain incompletely understood. Here, we identify a 44 bp transcriptional enhancer that is sufficient to direct expression specifically and exclusively to the developing vascular endothelium. This enhancer is regulated by a composite cis-acting element, the FOX:ETS motif, which is bound and synergistically activated by Forkhead and Ets transcription factors. We demonstrate that coexpression of the Forkhead protein FoxC2 and the Ets protein Etv2 induces ectopic expression of vascular genes in Xenopus embryos, and that combinatorial knockdown of the orthologous genes in zebrafish embryos disrupts vascular development. Finally, we show that FOX:ETS motifs are present in many known endothelial-specific enhancers and that this motif is an efficient predictor of endothelial enhancers in the human genome.


Asunto(s)
Elementos de Facilitación Genéticos , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Proto-Oncogénicas c-ets/metabolismo , Animales , Vasos Sanguíneos/embriología , Embrión de Mamíferos/citología , Embrión no Mamífero/metabolismo , Endotelio/embriología , Fibroblastos/metabolismo , Humanos , Ratones , Xenopus , Pez Cebra
6.
Circ Res ; 127(12): 1502-1518, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33044128

RESUMEN

RATIONALE: Cardiac pacemaker cells (PCs) in the sinoatrial node (SAN) have a distinct gene expression program that allows them to fire automatically and initiate the heartbeat. Although critical SAN transcription factors, including Isl1 (Islet-1), Tbx3 (T-box transcription factor 3), and Shox2 (short-stature homeobox protein 2), have been identified, the cis-regulatory architecture that governs PC-specific gene expression is not understood, and discrete enhancers required for gene regulation in the SAN have not been identified. OBJECTIVE: To define the epigenetic profile of PCs using comparative ATAC-seq (assay for transposase-accessible chromatin with sequencing) and to identify novel enhancers involved in SAN gene regulation, development, and function. METHODS AND RESULTS: We used ATAC-seq on sorted neonatal mouse SAN to compare regions of accessible chromatin in PCs and right atrial cardiomyocytes. PC-enriched assay for transposase-accessible chromatin peaks, representing candidate SAN regulatory elements, were located near established SAN genes and were enriched for distinct sets of TF (transcription factor) binding sites. Among several novel SAN enhancers that were experimentally validated using transgenic mice, we identified a 2.9-kb regulatory element at the Isl1 locus that was active specifically in the cardiac inflow at embryonic day 8.5 and throughout later SAN development and maturation. Deletion of this enhancer from the genome of mice resulted in SAN hypoplasia and sinus arrhythmias. The mouse SAN enhancer also directed reporter activity to the inflow tract in developing zebrafish hearts, demonstrating deep conservation of its upstream regulatory network. Finally, single nucleotide polymorphisms in the human genome that occur near the region syntenic to the mouse enhancer exhibit significant associations with resting heart rate in human populations. CONCLUSIONS: (1) PCs have distinct regions of accessible chromatin that correlate with their gene expression profile and contain novel SAN enhancers, (2) cis-regulation of Isl1 specifically in the SAN depends upon a conserved SAN enhancer that regulates PC development and SAN function, and (3) a corresponding human ISL1 enhancer may regulate human SAN function.


Asunto(s)
Arritmia Sinusal/metabolismo , Relojes Biológicos , Secuenciación de Inmunoprecipitación de Cromatina , Elementos de Facilitación Genéticos , Frecuencia Cardíaca , Proteínas con Homeodominio LIM/metabolismo , Nodo Sinoatrial/metabolismo , Factores de Transcripción/metabolismo , Potenciales de Acción , Animales , Arritmia Sinusal/genética , Arritmia Sinusal/fisiopatología , Epigénesis Genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Humanos , Proteínas con Homeodominio LIM/genética , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Polimorfismo de Nucleótido Simple , Nodo Sinoatrial/fisiopatología , Factores de Tiempo , Factores de Transcripción/genética , Pez Cebra/genética , Pez Cebra/metabolismo
7.
Circ Res ; 127(12): 1522-1535, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33040635

RESUMEN

RATIONALE: The development and function of the pacemaker cardiomyocytes of the sinoatrial node (SAN), the leading pacemaker of the heart, are tightly controlled by a conserved network of transcription factors, including TBX3 (T-box transcription factor 3), ISL1 (ISL LIM homeobox 1), and SHOX2 (short stature homeobox 2). Yet, the regulatory DNA elements (REs) controlling target gene expression in the SAN pacemaker cells have remained undefined. OBJECTIVE: Identification of the regulatory landscape of human SAN-like pacemaker cells and functional assessment of SAN-specific REs potentially involved in pacemaker cell gene regulation. METHODS AND RESULTS: We performed Assay for Transposase-Accessible Chromatin using sequencing on human pluripotent stem cell-derived SAN-like pacemaker cells and ventricle-like cells and identified thousands of putative REs specific for either human cell type. We validated pacemaker cell-specific elements in the SHOX2 and TBX3 loci. CRISPR-mediated homozygous deletion of the mouse ortholog of a noncoding region with candidate pacemaker-specific REs in the SHOX2 locus resulted in selective loss of Shox2 expression from the developing SAN and embryonic lethality. Putative pacemaker-specific REs were identified up to 1 Mbp upstream of TBX3 in a region close to MED13L harboring variants associated with heart rate recovery after exercise. The orthologous region was deleted in mice, which resulted in selective loss of expression of Tbx3 from the SAN and (cardiac) ganglia and in neonatal lethality. Expression of Tbx3 was maintained in other tissues including the atrioventricular conduction system, lungs, and liver. Heterozygous adult mice showed increased SAN recovery times after pacing. The human REs harboring the associated variants robustly drove expression in the SAN of transgenic mouse embryos. CONCLUSIONS: We provided a genome-wide collection of candidate human pacemaker-specific REs, including the loci of SHOX2, TBX3, and ISL1, and identified a link between human genetic variants influencing heart rate recovery after exercise and a variant RE with highly conserved function, driving SAN expression of TBX3.


Asunto(s)
Relojes Biológicos , Elementos de Facilitación Genéticos , Frecuencia Cardíaca , Miocitos Cardíacos/metabolismo , Nodo Sinoatrial/metabolismo , Proteínas de Dominio T Box/metabolismo , Potenciales de Acción , Animales , Línea Celular , Epigénesis Genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Ratones Transgénicos , Mutación , Proteínas de Dominio T Box/genética , Pez Cebra
8.
Nature ; 532(7598): 201-6, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-27049946

RESUMEN

How tissue regeneration programs are triggered by injury has received limited research attention. Here we investigate the existence of enhancer regulatory elements that are activated in regenerating tissue. Transcriptomic analyses reveal that leptin b (lepb) is highly induced in regenerating hearts and fins of zebrafish. Epigenetic profiling identified a short DNA sequence element upstream and distal to lepb that acquires open chromatin marks during regeneration and enables injury-dependent expression from minimal promoters. This element could activate expression in injured neonatal mouse tissues and was divisible into tissue-specific modules sufficient for expression in regenerating zebrafish fins or hearts. Simple enhancer-effector transgenes employing lepb-linked sequences upstream of pro- or anti-regenerative factors controlled the efficacy of regeneration in zebrafish. Our findings provide evidence for 'tissue regeneration enhancer elements' (TREEs) that trigger gene expression in injury sites and can be engineered to modulate the regenerative potential of vertebrate organs.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Especificidad de Órganos/genética , Regeneración/genética , Regeneración/fisiología , Cicatrización de Heridas/genética , Pez Cebra/genética , Pez Cebra/fisiología , Acetilación , Aletas de Animales/lesiones , Aletas de Animales/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular , Ensamble y Desensamble de Cromatina/genética , Epigénesis Genética/genética , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Corazón , Histonas/química , Histonas/metabolismo , Leptina/biosíntesis , Leptina/genética , Lisina/metabolismo , Masculino , Ratones , Miocitos Cardíacos/citología , Regiones Promotoras Genéticas/genética , Transgenes/genética , Proteínas de Pez Cebra/genética
9.
Dev Biol ; 445(2): 170-177, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30521808

RESUMEN

MEF2C is a member of the highly conserved MEF2 family of transcription factors and is a key regulator of cardiovascular development. In mice, Mef2c is expressed in the developing heart and vasculature, including the endothelium. Loss of Mef2c function in germline knockout mice leads to early embryonic demise and profound developmental abnormalities in the cardiovascular system. Previous attempts to uncover the cause of embryonic lethality by specifically disrupting Mef2c function in the heart or vasculature failed to recapitulate the global Mef2c knockout phenotype and instead resulted in relatively minor defects that did not compromise viability or result in significant cardiovascular defects. However, previous studies examined the requirement of Mef2c in the myocardial and endothelial lineages using Cre lines that begin to be expressed after the expression of Mef2c has already commenced. Here, we tested the requirement of Mef2c in the myocardial and endothelial lineages using conditional knockout approaches in mice with Cre lines that deleted Mef2c prior to onset of its expression in embryonic development. We found that deletion of Mef2c in the early myocardial lineage using Nkx2-5Cre resulted in cardiac and vascular abnormalities that were indistinguishable from the defects in the global Mef2c knockout. In contrast, early deletion of Mef2c in the vascular endothelium using an Etv2::Cre line active prior to the onset of Mef2c expression resulted in viable offspring that were indistinguishable from wild type controls with no overt defects in vascular development, despite nearly complete early deletion of Mef2c in the vascular endothelium. Thus, these studies support the idea that the requirement of MEF2C for vascular development is secondary to its requirement in the heart and suggest that the observed failure in vascular remodeling in Mef2c knockout mice results from defective heart function.


Asunto(s)
Sistema Cardiovascular/embriología , Animales , Fenómenos Fisiológicos Cardiovasculares/genética , Endotelio Vascular/anomalías , Endotelio Vascular/embriología , Femenino , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/genética , Factores de Transcripción MEF2/deficiencia , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Organogénesis/genética , Organogénesis/fisiología , Embarazo
10.
Development ; 144(7): 1235-1241, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28351867

RESUMEN

Enhancers frequently contain multiple binding sites for the same transcription factor. These homotypic binding sites often exhibit synergy, whereby the transcriptional output from two or more binding sites is greater than the sum of the contributions of the individual binding sites alone. Although this phenomenon is frequently observed, the mechanistic basis for homotypic binding site synergy is poorly understood. Here, we identify a bona fide cardiac-specific Prkaa2 enhancer that is synergistically activated by homotypic MEF2 binding sites. We show that two MEF2 sites in the enhancer function cooperatively due to bridging of the MEF2C-bound sites by the SAP domain-containing co-activator protein myocardin, and we show that paired sites buffer the enhancer from integration site-dependent effects on transcription in vivo Paired MEF2 sites are prevalent in cardiac enhancers, suggesting that this might be a common mechanism underlying synergy in the control of cardiac gene expression in vivo.


Asunto(s)
Factores de Transcripción MEF2/metabolismo , Miocardio/metabolismo , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Transcripción Genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Elementos de Facilitación Genéticos , Ratones Transgénicos , Multimerización de Proteína
11.
Development ; 143(5): 774-9, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26811383

RESUMEN

Congenital heart defects are the most common birth defects in humans, and those that affect the proper alignment of the outflow tracts and septation of the ventricles are a highly significant cause of morbidity and mortality in infants. A late differentiating population of cardiac progenitors, referred to as the anterior second heart field (AHF), gives rise to the outflow tract and the majority of the right ventricle and provides an embryological context for understanding cardiac outflow tract alignment and membranous ventricular septal defects. However, the transcriptional pathways controlling AHF development and their roles in congenital heart defects remain incompletely elucidated. Here, we inactivated the gene encoding the transcription factor MEF2C in the AHF in mice. Loss of Mef2c function in the AHF results in a spectrum of outflow tract alignment defects ranging from overriding aorta to double-outlet right ventricle and dextro-transposition of the great arteries. We identify Tdgf1, which encodes a Nodal co-receptor (also known as Cripto), as a direct transcriptional target of MEF2C in the outflow tract via an AHF-restricted Tdgf1 enhancer. Importantly, both the MEF2C and TDGF1 genes are associated with congenital heart defects in humans. Thus, these studies establish a direct transcriptional pathway between the core cardiac transcription factor MEF2C and the human congenital heart disease gene TDGF1. Moreover, we found a range of outflow tract alignment defects resulting from a single genetic lesion, supporting the idea that AHF-derived outflow tract alignment defects may constitute an embryological spectrum rather than distinct anomalies.


Asunto(s)
Factor de Crecimiento Epidérmico/fisiología , Regulación del Desarrollo de la Expresión Génica , Glicoproteínas de Membrana/fisiología , Proteínas de Neoplasias/fisiología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Factor de Crecimiento Epidérmico/genética , Femenino , Eliminación de Gen , Corazón/embriología , Cardiopatías Congénitas/genética , Defectos del Tabique Interventricular/genética , Ventrículos Cardíacos , Humanos , Hibridación in Situ , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/fisiología , Masculino , Glicoproteínas de Membrana/genética , Ratones , Morfogénesis/genética , Proteínas de Neoplasias/genética , Organogénesis , Análisis de Secuencia de ARN , Distribución Tisular , Transcripción Genética , Transposición de los Grandes Vasos/genética
12.
Proc Natl Acad Sci U S A ; 113(40): 11237-11242, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27647901

RESUMEN

Zebrafish have a remarkable capacity to regenerate their heart. Efficient replenishment of lost tissues requires the activation of different cell types including the epicardium and endocardium. A complex set of processes is subsequently needed to support cardiomyocyte repopulation. Previous studies have identified important determinants of heart regeneration; however, to date, how revascularization of the damaged area happens remains unknown. Here, we show that angiogenic sprouting into the injured area starts as early as 15 h after injury. To analyze the role of vegfaa in heart regeneration, we used vegfaa mutants rescued to adulthood by vegfaa mRNA injections at the one-cell stage. Surprisingly, vegfaa mutants develop coronaries and revascularize after injury. As a possible explanation for these observations, we find that vegfaa mutant hearts up-regulate the expression of potentially compensating genes. Therefore, to overcome the lack of a revascularization phenotype in vegfaa mutants, we generated fish expressing inducible dominant negative Vegfaa. These fish displayed minimal revascularization of the damaged area. In the absence of fast angiogenic revascularization, cardiomyocyte proliferation did not occur, and the heart failed to regenerate, retaining a fibrotic scar. Hence, our data show that a fast endothelial invasion allows efficient revascularization of the injured area, which is necessary to support replenishment of new tissue and achieve efficient heart regeneration. These findings revisit the model where neovascularization is considered to happen concomitant with the formation of new muscle. Our work also paves the way for future studies designed to understand the molecular mechanisms that regulate fast revascularization.


Asunto(s)
Corazón/fisiopatología , Revascularización Miocárdica , Regeneración/fisiología , Pez Cebra/fisiología , Animales , Biomarcadores/metabolismo , Proliferación Celular , Supervivencia Celular , Vasos Coronarios/patología , Regulación del Desarrollo de la Expresión Génica , Respuesta al Choque Térmico , Mutación/genética , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , Pericardio/patología , Conducto Torácico/patología , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
13.
Development ; 142(16): 2775-80, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26160899

RESUMEN

Endothelin signaling is essential for neural crest development, and dysregulated Endothelin signaling is associated with several neural crest-related disorders, including Waardenburg and other syndromes. However, despite the crucial roles of this pathway in neural crest development and disease, the transcriptional effectors directly activated by Endothelin signaling during neural crest development remain incompletely elucidated. Here, we establish that the MADS box transcription factor MEF2C is an immediate downstream transcriptional target and effector of Endothelin signaling in the neural crest. We show that Endothelin signaling activates Mef2c expression in the neural crest through a conserved enhancer in the Mef2c locus and that CRISPR-mediated deletion of this Mef2c neural crest enhancer from the mouse genome abolishes Endothelin induction of Mef2c expression. Moreover, we demonstrate that Endothelin signaling activates neural crest expression of Mef2c by de-repressing MEF2C activity through a Calmodulin-CamKII-histone deacetylase signaling cascade. Thus, these findings identify a MEF2C-dependent, positive-feedback mechanism for Endothelin induction and establish MEF2C as an immediate transcriptional effector and target of Endothelin signaling in the neural crest.


Asunto(s)
Endotelinas/metabolismo , Retroalimentación Fisiológica/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Cresta Neural/fisiología , Transducción de Señal/fisiología , Animales , Galactósidos , Hibridación in Situ , Indoles , Factores de Transcripción MEF2/metabolismo , Ratones , Ratones Transgénicos , Cresta Neural/metabolismo , beta-Galactosidasa
14.
Dev Biol ; 402(1): 3-16, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25794678

RESUMEN

Neural crest cells (NCC) comprise a multipotent, migratory stem cell and progenitor population that gives rise to numerous cell and tissue types within a developing embryo, including craniofacial bone and cartilage, neurons and glia of the peripheral nervous system, and melanocytes within the skin. Here we describe two novel stable transgenic mouse lines suitable for lineage tracing and analysis of gene function in NCC. Firstly, using the F10N enhancer of the Mef2c gene (Mef2c-F10N) linked to LacZ, we generated transgenic mice (Mef2c-F10N-LacZ) that express LacZ in the majority, if not all migrating NCC that delaminate from the neural tube. Mef2c-F10N-LacZ then continues to be expressed primarily in neurogenic, gliogenic and melanocytic NCC and their derivatives, but not in ectomesenchymal derivatives. Secondly, we used the same Mef2c-F10N enhancer together with Cre recombinase to generate transgenic mice (Mef2c-F10N-Cre) that can be used to indelibly label, or alter gene function in, migrating NCC and their derivatives. At early stages of development, Mef2c-F10N-LacZ and Mef2c-F10N-Cre label NCC in a pattern similar to Wnt1-Cre mice, with the exception that Mef2c-F10N-LacZ and Mef2c-F10N-Cre specifically label NCC that have delaminated from the neural plate, while premigratory NCC are not labeled. Thus, our Mef2c-F10N-LacZ and Mef2c-F10N-Cre transgenic mice provide new resources for tracing migratory NCC and analyzing gene function in migrating and differentiating NCC independently of NCC formation.


Asunto(s)
Elementos de Facilitación Genéticos , Integrasas/genética , Operón Lac , Ratones Transgénicos , Cresta Neural/citología , Animales , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Pollos , Regulación del Desarrollo de la Expresión Génica , Genotipo , Humanos , Integrasas/metabolismo , Melanocitos/citología , Mesodermo/metabolismo , Ratones , Cresta Neural/metabolismo , Neuronas/metabolismo , Conejos , Ratas , Xenopus , Pez Cebra
15.
Evol Dev ; 18(1): 31-40, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26086993

RESUMEN

Mammalian dental morphology is under strong evolutionary pressure because of its importance for mastication and diet. While the mechanisms underlying tooth development have been widely studied in model organisms, the role of genetic regulatory elements in patterning the different elements of the occlusal surface and crown height across species is not well understood. Previous studies showed that Fibroblast Growth Factor (FGF) genes are important regulators of tooth development that influence morphological variation. We hypothesized that inter-specific variation in rodent dental morphology could be governed by nucleotide variation in genetic regulatory elements that modulate the spatial and temporal expression of the genes encoding FGF signaling molecules. In this study, we compared the variation in dental morphology across nine taxa of rodents to the variation in sequences of non-coding evolutionary conserved regions (ECRs) of Fgf3, 4, 8, 9, and 10. We correlated the variation in molar tooth cusp shape and the evolution of high molar crowns (hypsodonty) to the patterns of sequence variation in two ECRs, Fgf10ECR3, and Fgf9ECR1, respectively. By conducting luciferase and electrophoretic mobility shift assays, we determined that these ECRs could function as enhancers. These data suggest that emergence of hypsodonty and occlusal cusp patterning may have happened through the evolutionary changes in enhancers, such as Fgf9ECR1 and Fgf10ECR3, which affected the expression of major signaling molecules involved in tooth development.


Asunto(s)
Evolución Biológica , Factores de Crecimiento de Fibroblastos/genética , Diente/fisiología , Vertebrados/genética , Animales , Secuencia Conservada , Diente Molar/anatomía & histología , Diente Molar/fisiología , Filogenia , Diente/anatomía & histología , Vertebrados/clasificación , Vertebrados/fisiología
17.
Proc Natl Acad Sci U S A ; 110(44): 17921-6, 2013 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-24127591

RESUMEN

Chromatin-based functional genomic analyses and genomewide association studies (GWASs) together implicate enhancers as critical elements influencing gene expression and risk for common diseases. Here, we performed systematic chromatin and transcriptome profiling in human pancreatic islets. Integrated analysis of islet data with those from nine cell types identified specific and significant enrichment of type 2 diabetes and related quantitative trait GWAS variants in islet enhancers. Our integrated chromatin maps reveal that most enhancers are short (median = 0.8 kb). Each cell type also contains a substantial number of more extended (≥ 3 kb) enhancers. Interestingly, these stretch enhancers are often tissue-specific and overlap locus control regions, suggesting that they are important chromatin regulatory beacons. Indeed, we show that (i) tissue specificity of enhancers and nearby gene expression increase with enhancer length; (ii) neighborhoods containing stretch enhancers are enriched for important cell type-specific genes; and (iii) GWAS variants associated with traits relevant to a particular cell type are more enriched in stretch enhancers compared with short enhancers. Reporter constructs containing stretch enhancer sequences exhibited tissue-specific activity in cell culture experiments and in transgenic mice. These results suggest that stretch enhancers are critical chromatin elements for coordinating cell type-specific regulatory programs and that sequence variation in stretch enhancers affects risk of major common human diseases.


Asunto(s)
Diferenciación Celular/fisiología , Cromatina/fisiología , Diabetes Mellitus Tipo 2/fisiopatología , Elementos de Facilitación Genéticos/genética , Epigenómica/métodos , Regulación de la Expresión Génica/fisiología , Células Secretoras de Insulina/metabolismo , Animales , Inmunoprecipitación de Cromatina , Diabetes Mellitus Tipo 2/genética , Elementos de Facilitación Genéticos/fisiología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Estudio de Asociación del Genoma Completo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Células Secretoras de Insulina/fisiología , Luciferasas , Ratones , Ratones Transgénicos
18.
Dev Biol ; 395(2): 379-389, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25179465

RESUMEN

Endothelin-converting enzyme-1 (Ece-1), a crucial component of the Endothelin signaling pathway, is required for embryonic development and is an important regulator of vascular tone, yet the transcriptional regulation of the ECE1 gene has remained largely unknown. Here, we define the activity and regulation of an enhancer from the human ECE1 locus in vivo. The enhancer identified here becomes active in endothelial progenitor cells shortly after their initial specification and is dependent on a conserved FOX:ETS motif, a composite binding site for Forkhead transcription factors and the Ets transcription factor Etv2, for activity in vivo. The ECE1 FOX:ETS motif is bound and cooperatively activated by FoxC2 and Etv2, but unlike other described FOX:ETS-dependent enhancers, ECE1 enhancer activity becomes restricted to arterial endothelium and endocardium by embryonic day 9.5 in transgenic mouse embryos. The ECE1 endothelial enhancer also contains an evolutionarily-conserved, consensus SOX binding site, which is required for activity in transgenic mouse embryos. Importantly, the ECE1 SOX site is bound and activated by Sox17, a transcription factor involved in endothelial cell differentiation and an important regulator of arterial identity. Moreover, the ECE1 enhancer is cooperatively activated by the combinatorial action of FoxC2, Etv2, and Sox17. Although Sox17 is required for arterial identity, few direct transcriptional targets have been identified in endothelial cells. Thus, this work has important implications for our understanding of endothelial specification and arterial subspecification.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Endocardio/embriología , Endotelio Vascular/embriología , Factores de Transcripción Forkhead/metabolismo , Metaloendopeptidasas/metabolismo , Factores de Transcripción SOXF/metabolismo , Factores de Transcripción/metabolismo , Animales , Ácido Aspártico Endopeptidasas/genética , Clonación Molecular , Cartilla de ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Endocardio/metabolismo , Enzimas Convertidoras de Endotelina , Endotelio Vascular/metabolismo , Elementos de Facilitación Genéticos/genética , Técnica del Anticuerpo Fluorescente , Galactósidos , Humanos , Indoles , Metaloendopeptidasas/genética , Ratones , Ratones Transgénicos , Mutagénesis , Factores de Transcripción SOX/metabolismo
19.
Dev Biol ; 393(2): 245-254, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25050930

RESUMEN

Coordinated contraction of the heart is essential for survival and is regulated by the cardiac conduction system. Contraction of ventricular myocytes is controlled by the terminal part of the conduction system known as the Purkinje fiber network. Lineage analyses in chickens and mice have established that the Purkinje fibers of the peripheral ventricular conduction system arise from working myocytes during cardiac development. It has been proposed, based primarily on gain-of-function studies, that Endothelin signaling is responsible for myocyte-to-Purkinje fiber transdifferentiation during avian heart development. However, the role of Endothelin signaling in mammalian conduction system development is less clear, and the development of the cardiac conduction system in mice lacking Endothelin signaling has not been previously addressed. Here, we assessed the specification of the cardiac conduction system in mouse embryos lacking all Endothelin signaling. We found that mouse embryos that were homozygous null for both ednra and ednrb, the genes encoding the two Endothelin receptors in mice, were born at predicted Mendelian frequency and had normal specification of the cardiac conduction system and apparently normal electrocardiograms with normal QRS intervals. In addition, we found that ednra expression within the heart was restricted to the myocardium while ednrb expression in the heart was restricted to the endocardium and coronary endothelium. By establishing that ednra and ednrb are expressed in distinct compartments within the developing mammalian heart and that Endothelin signaling is dispensable for specification and function of the cardiac conduction system, this work has important implications for our understanding of mammalian cardiac development.


Asunto(s)
Endotelinas/metabolismo , Contracción Miocárdica/fisiología , Ramos Subendocárdicos/embriología , Receptores de Endotelina/genética , Animales , Diferenciación Celular , Transdiferenciación Celular , Conexina 43/biosíntesis , Conexinas/biosíntesis , Endocardio/metabolismo , Endotelio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Organogénesis , Ramos Subendocárdicos/fisiología , Receptores de Endotelina/biosíntesis , Transducción de Señal , Proteína alfa-5 de Unión Comunicante
20.
Nature ; 461(7260): 95-8, 2009 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-19727199

RESUMEN

The emergence of terrestrial life witnessed the need for more sophisticated circulatory systems. This has evolved in birds, mammals and crocodilians into complete septation of the heart into left and right sides, allowing separate pulmonary and systemic circulatory systems, a key requirement for the evolution of endothermy. However, the evolution of the amniote heart is poorly understood. Reptilian hearts have been the subject of debate in the context of the evolution of cardiac septation: do they possess a single ventricular chamber or two incompletely septated ventricles? Here we examine heart development in the red-eared slider turtle, Trachemys scripta elegans (a chelonian), and the green anole, Anolis carolinensis (a squamate), focusing on gene expression in the developing ventricles. Both reptiles initially form a ventricular chamber that homogenously expresses the T-box transcription factor gene Tbx5. In contrast, in birds and mammals, Tbx5 is restricted to left ventricle precursors. In later stages, Tbx5 expression in the turtle (but not anole) heart is gradually restricted to a distinct left ventricle, forming a left-right gradient. This suggests that Tbx5 expression was refined during evolution to pattern the ventricles. In support of this hypothesis, we show that loss of Tbx5 in the mouse ventricle results in a single chamber lacking distinct identity, indicating a requirement for Tbx5 in septation. Importantly, misexpression of Tbx5 throughout the developing myocardium to mimic the reptilian expression pattern also results in a single mispatterned ventricular chamber lacking septation. Thus ventricular septation is established by a steep and correctly positioned Tbx5 gradient. Our findings provide a molecular mechanism for the evolution of the amniote ventricle, and support the concept that altered expression of developmental regulators is a key mechanism of vertebrate evolution.


Asunto(s)
Evolución Molecular , Corazón/embriología , Lagartos/embriología , Tortugas/embriología , Animales , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica , Corazón/anatomía & histología , Lagartos/anatomía & histología , Lagartos/genética , Ratones , Organogénesis , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Tortugas/anatomía & histología , Tortugas/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda