Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33443158

RESUMEN

The sinus node (SAN) is the primary pacemaker of the human heart, and abnormalities in its structure or function cause sick sinus syndrome, the most common reason for electronic pacemaker implantation. Here we report that transcription factor GATA6, whose mutations in humans are linked to arrhythmia, is highly expressed in the SAN and its haploinsufficiency in mice results in hypoplastic SANs and rhythm abnormalities. Cell-specific deletion reveals a requirement for GATA6 in various SAN lineages. Mechanistically, GATA6 directly activates key regulators of the SAN genetic program in conduction and nonconduction cells, such as TBX3 and EDN1, respectively. The data identify GATA6 as an important regulator of the SAN and provide a molecular basis for understanding the conduction abnormalities associated with GATA6 mutations in humans. They also suggest that GATA6 may be a potential modifier of the cardiac pacemaker.


Asunto(s)
Factor de Transcripción GATA6/metabolismo , Frecuencia Cardíaca/fisiología , Nodo Sinoatrial/embriología , Animales , Arritmias Cardíacas/fisiopatología , Diferenciación Celular/genética , Factor de Transcripción GATA6/genética , Regulación del Desarrollo de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organogénesis , Nodo Sinoatrial/fisiología , Proteínas de Dominio T Box/genética
2.
iScience ; 23(3): 100959, 2020 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-32179481

RESUMEN

Defining the pathways that control cardiac development facilitates understanding the pathogenesis of congenital heart disease. Herein, we identify enrichment of a Cullin5 Ub ligase key subunit, Asb2, in myocardial progenitors and differentiated cardiomyocytes. Using two conditional murine knockouts, Nkx+/Cre.Asb2fl/fl and AHF-Cre.Asb2fl/fl, and tissue clarifying technique, we reveal Asb2 requirement for embryonic survival and complete heart looping. Deletion of Asb2 results in upregulation of its target Filamin A (Flna), and concurrent Flna deletion partially rescues embryonic lethality. Conditional AHF-Cre.Asb2 knockouts harboring one Flna allele have double outlet right ventricle (DORV), which is rescued by biallelic Flna excision. Transcriptomic and immunofluorescence analyses identify Tgfß/Smad as downstream targets of Asb2/Flna. Finally, using CRISPR/Cas9 genome editing, we demonstrate Asb2 requirement for human cardiomyocyte differentiation suggesting a conserved mechanism between mice and humans. Collectively, our study provides deeper mechanistic understanding of the role of the ubiquitin proteasome system in cardiac development and suggests a previously unidentified murine model for DORV.

3.
Circ Res ; 123(9): 1066-1079, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30355156

RESUMEN

RATIONALE: Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are a readily available, robustly reproducible, and physiologically appropriate human cell source for cardiac disease modeling, drug discovery, and toxicity screenings in vitro. However, unlike adult myocardial cells in vivo, hPSC-CMs cultured in vitro maintain an immature metabolic phenotype, where majority of ATP is produced through aerobic glycolysis instead of oxidative phosphorylation in the mitochondria. Little is known about the underlying signaling pathways controlling hPSC-CMs' metabolic and functional maturation. OBJECTIVE: To define the molecular pathways controlling cardiomyocytes' metabolic pathway selections and improve cardiomyocyte metabolic and functional maturation. METHODS AND RESULTS: We cultured hPSC-CMs in different media compositions including glucose-containing media, glucose-containing media supplemented with fatty acids, and glucose-free media with fatty acids as the primary carbon source. We found that cardiomyocytes cultured in the presence of glucose used primarily aerobic glycolysis and aberrantly upregulated HIF1α (hypoxia-inducible factor 1α) and its downstream target lactate dehydrogenase A. Conversely, glucose deprivation promoted oxidative phosphorylation and repressed HIF1α. Small molecule inhibition of HIF1α or lactate dehydrogenase A resulted in a switch from aerobic glycolysis to oxidative phosphorylation. Likewise, siRNA inhibition of HIF1α stimulated oxidative phosphorylation while inhibiting aerobic glycolysis. This metabolic shift was accompanied by an increase in mitochondrial content and cellular ATP levels. Furthermore, functional gene expressions, sarcomere length, and contractility were improved by HIF1α/lactate dehydrogenase A inhibition. CONCLUSIONS: We show that under standard culture conditions, the HIF1α-lactate dehydrogenase A axis is aberrantly upregulated in hPSC-CMs, preventing their metabolic maturation. Chemical or siRNA inhibition of this pathway results in an appropriate metabolic shift from aerobic glycolysis to oxidative phosphorylation. This in turn improves metabolic and functional maturation of hPSC-CMs. These findings provide key insight into molecular control of hPSC-CMs' metabolism and may be used to generate more physiologically mature cardiomyocytes for drug screening, disease modeling, and therapeutic purposes.


Asunto(s)
Aminoquinolinas/farmacología , Diferenciación Celular/efectos de los fármacos , Disulfuros/farmacología , Metabolismo Energético/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Alcaloides Indólicos/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , L-Lactato Deshidrogenasa/antagonistas & inhibidores , Mitocondrias Cardíacas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Sulfonamidas/farmacología , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Glucólisis/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células Madre Pluripotentes Inducidas/enzimología , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/enzimología , Mitocondrias Cardíacas/genética , Miocitos Cardíacos/enzimología , Fosforilación Oxidativa/efectos de los fármacos , Fenotipo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos
4.
Hum Mol Genet ; 26(5): 942-954, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28164238

RESUMEN

TBX5, a member of the T-box family of transcription factors, is a dosage sensitive regulator of heart development. Mutations in TBX5 are responsible for Holt-Oram Syndrome, an autosomal dominant disease with variable and partially penetrant cardiac defects suggestive of the existence of genetic and environmental modifiers. KLF13, a member of the Krüppel-like family of zinc finger proteins is co-expressed with TBX5 in several cardiac cells including atrial cardiomyocytes and cells of the interatrial septum. We report that KLF13 interacts physically and functionally with TBX5 to synergistically activate transcription of cardiac genes. We show that TBX5 contacts KLF13 via its T-domain and find that several disease-causing mutations therein have decreased KLF13 interaction. Whereas Klf13 heterozygote mice have no detectable cardiac defects, loss of a Klf13 allele in Tbx5 heterozygote mice significantly increases the penetrance of TBX5-dependent cardiac abnormalities including atrial, atrial-ventricular and ventricular septal defects. The results reveal for the first time combinatorial interaction between a T-box protein and a KLF family member and its importance for heart and possibly other organ development. The data also suggest that, in human, KLF13 may be a genetic modifier of the Holt-Oram Syndrome gene TBX5.


Asunto(s)
Anomalías Múltiples/genética , Proteínas de Ciclo Celular/genética , Atrios Cardíacos/metabolismo , Cardiopatías Congénitas/genética , Defectos del Tabique Interatrial/genética , Factores de Transcripción de Tipo Kruppel/genética , Deformidades Congénitas de las Extremidades Inferiores/genética , Proteínas Represoras/genética , Proteínas de Dominio T Box/genética , Deformidades Congénitas de las Extremidades Superiores/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Animales , Proteínas de Ciclo Celular/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Atrios Cardíacos/patología , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/patología , Defectos del Tabique Interatrial/metabolismo , Defectos del Tabique Interatrial/patología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Heterocigoto , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Deformidades Congénitas de las Extremidades Inferiores/metabolismo , Deformidades Congénitas de las Extremidades Inferiores/patología , Ratones , Mutación , Unión Proteica , Dominios Proteicos/genética , Mapas de Interacción de Proteínas/genética , Proteínas Represoras/metabolismo , Proteínas de Dominio T Box/metabolismo , Activación Transcripcional/genética , Deformidades Congénitas de las Extremidades Superiores/metabolismo , Deformidades Congénitas de las Extremidades Superiores/patología
5.
J Biol Chem ; 290(11): 6844-56, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25623069

RESUMEN

TBX5 is the gene mutated in Holt-Oram syndrome, an autosomal dominant disorder with complex heart and limb deformities. Its protein product is a member of the T-box family of transcription factors and an evolutionarily conserved dosage-sensitive regulator of heart and limb development. Understanding TBX5 regulation is therefore of paramount importance. Here we uncover the existence of novel exons and provide evidence that TBX5 activity may be extensively regulated through alternative splicing to produce protein isoforms with differing N- and C-terminal domains. These isoforms are also present in human heart, indicative of an evolutionarily conserved regulatory mechanism. The newly identified isoforms have different transcriptional properties and can antagonize TBX5a target gene activation. Droplet Digital PCR as well as immunohistochemistry with isoform-specific antibodies reveal differential as well as overlapping expression domains. In particular, we find that the predominant isoform in skeletal myoblasts is Tbx5c, and we show that it is dramatically up-regulated in differentiating myotubes and is essential for myotube formation. Mechanistically, TBX5c antagonizes TBX5a activation of pro-proliferative signals such as IGF-1, FGF-10, and BMP4. The results provide new insight into Tbx5 regulation and function that will further our understanding of its role in health and disease. The finding of new exons in the Tbx5 locus may also be relevant to mutational screening especially in the 30% of Holt-Oram syndrome patients with no mutations in the known TBX5a exons.


Asunto(s)
Exones , Proteínas de Dominio T Box/análisis , Proteínas de Dominio T Box/genética , Anomalías Múltiples/genética , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Expresión Génica , Cardiopatías Congénitas/genética , Defectos del Tabique Interatrial/genética , Humanos , Deformidades Congénitas de las Extremidades Inferiores/genética , Ratones , Datos de Secuencia Molecular , Células Musculares/citología , Células Musculares/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Mutación , Miocardio/metabolismo , Miocardio/ultraestructura , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Alineación de Secuencia , Proteínas de Dominio T Box/metabolismo , Deformidades Congénitas de las Extremidades Superiores/genética
6.
Mech Dev ; 134: 31-41, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25241353

RESUMEN

GATA4-6 transcription factors regulate numerous aspects of development and homeostasis in multiple tissues of mesodermal and endodermal origin. In the heart, the best studied of these factors, GATA4, has multiple distinct roles in cardiac specification, differentiation, morphogenesis, hypertrophy and survival. To improve understanding of how GATA4 achieves its numerous roles in the heart, here we have focused on the carboxy-terminal domain and the residues required for interaction with cofactors FOG2 and Tbx5. We present evidence that the carboxy terminal region composed of amino acids 362-400 is essential for mediating cardiogenesis in Xenopus pluripotent explants and embryos. In contrast, the same region is not required for endoderm-inducing activity of GATA4. Further evidence is presented that the carboxy terminal cardiogenic region of GATA4 does not operate as a generic transcriptional activator. Potential mechanism of action of the carboxy terminal end of GATA4 is provided by the results showing physical and functional interaction with CDK4, including the enhancement of cardiogenic activity of GATA4 by CDK4. These results establish CDK4 as a GATA4 partner in cardiogenesis. The interactions of GATA4 with its other well described cofactors Tbx5 and FOG2 are known to be involved in heart morphogenesis, but their requirement for cardiac differentiation is unknown. We report that the mutations that disrupt interactions of GATA4 with Tbx5 and FOG2, G295S and V217G, respectively, do not impair cardiogenic activity of GATA4. These findings add support to the view that distinct roles of GATA4 in the heart are mediated by different determinants of the protein. Finally, we show that the rat GATA4 likely induces cardiogenesis cell autonomously or directly as it does not require activity of endodermal transcription factor Sox17, a GATA4 target gene that induces cardiogenesis non-cell autonomously.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/metabolismo , Factor de Transcripción GATA4/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Miocardio/metabolismo , Células 3T3 , Animales , Diferenciación Celular/genética , Quinasa 4 Dependiente de la Ciclina/genética , Factor de Transcripción GATA4/genética , Ratones , Miocitos Cardíacos/metabolismo , Organogénesis/genética , Ratas , Xenopus laevis
7.
Proc Natl Acad Sci U S A ; 111(4): 1415-20, 2014 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-24474767

RESUMEN

The G1 cyclins play a pivotal role in regulation of cell differentiation and proliferation. The mechanisms underlying their cell-specific roles are incompletely understood. Here, we show that a G1 cyclin, cyclin D2 (CycD2), enhances the activity of transcription factor GATA4, a key regulator of cardiomyocyte growth and differentiation. GATA4 recruits CycD2 to its target promoters, and their interaction results in synergistic activation of GATA-dependent transcription. This effect is specific to CycD2 because CycD1 is unable to potentiate activity of GATA4 and is CDK-independent. GATA4 physically interacts with CycD2 through a discreet N-terminal activation domain that is essential for the cardiogenic activity of GATA4. Human mutations in this domain that are linked to congenital heart disease interfere with CycD2-GATA4 synergy. Cardiogenesis assays in Xenopus embryos indicate that CycD2 enhances the cardiogenic function of GATA4. Together, our data uncover a role for CycD2 as a cardiogenic coactivator of GATA4 and suggest a paradigm for cell-specific effects of cyclin Ds.


Asunto(s)
Ciclina D2/fisiología , Factor de Transcripción GATA4/fisiología , Corazón/embriología , Organogénesis/fisiología , Secuencia de Aminoácidos , Animales , Ciclina D2/química , Humanos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Transactivadores/fisiología
8.
Am J Physiol Heart Circ Physiol ; 303(8): H1057-66, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22923619

RESUMEN

Transcription factor GATA4 is a key regulator of cardiomyocyte growth, and differentiation and 50% reduction in GATA4 levels results in hypoplastic hearts. Search for GATA4 targets/effectors revealed cyclin D(2) (CD2), a member of the D-type cyclins (D(1), D(2), and D(3)) that play a vital role in cell growth and differentiation as a direct transcriptional target and a mediator of GATA4 growth in postnatal cardiomyocytes. GATA4 associates with the CD2 promoter in cardiomyocytes and is sufficient to induce endogenous CD2 transcription and to dose-dependently activate the CD2 promoter in heterologous cells. Cardiomyocyte-specific overexpression of CD2 results in enhanced postnatal cardiac growth because of increased cardiomyocyte proliferation. When these transgenic mice are crossed with Gata4 heterozygote mice, they rescue the hypoplastic cardiac phenotype of Gata4(+/-) mice and enhance cardiomyocyte survival and heart function. The data uncover a role for CD2 in the postnatal heart as an effector of GATA4 in myocyte growth and survival. The finding that postnatal upregulation of a cell-cycle gene in GATA4 haplo-insufficient hearts may be protective opens new avenues for maintaining or restoring cardiac function in GATA4-dependent cardiac disease.


Asunto(s)
Cardiomegalia/fisiopatología , Ciclina D2/metabolismo , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Miocitos Cardíacos/fisiología , Animales , Apoptosis/fisiología , Antígenos CD2/genética , Antígenos CD2/metabolismo , Cardiomegalia/metabolismo , Proliferación Celular , Ciclina D2/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Células HEK293 , Haplotipos , Corazón/embriología , Corazón/fisiología , Humanos , Ratones , Ratones Transgénicos , Miocitos Cardíacos/citología , Células 3T3 NIH , Fenotipo , Ratas , Ratas Sprague-Dawley , Transcripción Genética/fisiología
9.
Proc Natl Acad Sci U S A ; 107(45): 19356-61, 2010 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-20974940

RESUMEN

In humans, septal defects are among the most prevalent congenital heart diseases, but their cellular and molecular origins are not fully understood. We report that transcription factor Tbx5 is present in a subpopulation of endocardial cells and that its deletion therein results in fully penetrant, dose-dependent atrial septal defects in mice. Increased apoptosis of endocardial cells lacking Tbx5, as well as neighboring TBX5-positive myocardial cells of the atrial septum through activation of endocardial NOS (Nos3), is the underlying mechanism of disease. Compound Tbx5 and Nos3 haploinsufficiency in mice worsens the cardiac phenotype. The data identify a pathway for endocardial cell survival and unravel a cell-autonomous role for Tbx5 therein. The finding that Nos3, a gene regulated by many congenital heart disease risk factors including stress and diabetes, interacts genetically with Tbx5 provides a molecular framework to understand gene-environment interaction in the setting of human birth defects.


Asunto(s)
Tabique Interatrial/citología , Endocardio/citología , Factor de Transcripción GATA4/fisiología , Cardiopatías/congénito , Óxido Nítrico Sintasa de Tipo III/fisiología , Proteínas de Dominio T Box/fisiología , Animales , Tabique Interatrial/patología , Supervivencia Celular , Endocardio/patología , Haploinsuficiencia , Cardiopatías Congénitas/etiología , Cardiopatías Congénitas/genética , Ratones , Fenotipo , Proteínas de Dominio T Box/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...