Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Tissue Res ; 391(1): 205-215, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36385586

RESUMEN

PINCH, an adaptor of focal adhesion complex, plays essential roles in multiple cellular processes and organogenesis. Here, we ablated PINCH1 or both of PINCH1 and PINCH2 in skeletal muscle progenitors using MyoD-Cre. Double ablation of PINCH1 and PINCH2 resulted in early postnatal lethality with reduced size of skeletal muscles and detachment of diaphragm muscles from the body wall. PINCH mutant myofibers failed to undergo multinucleation and exhibited disrupted sarcomere structures. The mutant myoblasts in culture were able to adhere to newly formed myotubes but impeded in cell fusion and subsequent sarcomere genesis and cytoskeleton organization. Consistent with this, expression of integrin ß1 and some cytoskeleton proteins and phosphorylation of ERK and AKT were significantly reduced in PINCH mutants. However, N-cadherin was correctly expressed at cell adhesion sites in PINCH mutant cells, suggesting that PINCH may play a direct role in myoblast fusion. Expression of MRF4, the most highly expressed myogenic factor at late stages of myogenesis, was abolished in PINCH mutants that could contribute to observed phenotypes. In addition, mice with PINCH1 being ablated in myogenic progenitors exhibited only mild centronuclear myopathic changes, suggesting a compensatory role of PINCH2 in myogenic differentiation. Our results revealed a critical role of PINCH proteins in myogenic differentiation.


Asunto(s)
Diferenciación Celular , Mioblastos Esqueléticos , Animales , Ratones , Adhesión Celular , Comunicación Celular , Adhesiones Focales/metabolismo , Músculo Esquelético/fisiología
2.
Am J Physiol Renal Physiol ; 315(2): F374-F385, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29638158

RESUMEN

Kidney development involves reciprocal and inductive interactions between the ureteric bud (UB) and surrounding metanephric mesenchyme. Signals from renal stromal lineages are essential for differentiation and patterning of renal epithelial and mesenchymal cell types and renal vasculogenesis; however, underlying mechanisms remain not fully understood. Integrin-linked kinase (ILK), a key component of integrin signaling pathway, plays an important role in kidney development. However, the role of ILK in renal stroma remains unknown. Here, we ablated ILK in renal stromal lineages using a platelet-derived growth factor receptor B ( Pdgfrb) -Cre mouse line, and the resulting Ilk mutant mice presented postnatal growth retardation and died within 3 wk of age with severe renal developmental defects. Pdgfrb-Cre;Ilk mutant kidneys exhibited a significant decrease in UB branching and disrupted collecting duct formation. From E16.5 onward, renal interstitium was disorganized, forming medullary interstitial pseudocysts. Pdgfrb-Cre;Ilk mutants exhibited renal vasculature mispatterning and impaired glomerular vascular differentiation. Impaired glial cell-derived neurotrophic factor/Ret and bone morphogenetic protein 7 signaling pathways were observed in Pdgfrb-Cre;Ilk mutant kidneys. Furthermore, phosphoproteomic and Western blot analyses revealed a significant dysregulation of a number of key signaling pathways required for kidney morphogenesis, including PI3K/AKT and MAPK/ERK in Pdgfrb-Cre;Ilk mutants. Our results revealed a critical requirement for ILK in renal-stromal and vascular development, as well as a noncell autonomous role of ILK in UB branching morphogenesis.


Asunto(s)
Riñón/enzimología , Enfermedades Renales Poliquísticas/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Células del Estroma/enzimología , Animales , Proteína Morfogenética Ósea 7/genética , Proteína Morfogenética Ósea 7/metabolismo , Diferenciación Celular , Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica , Predisposición Genética a la Enfermedad , Edad Gestacional , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Integrasas/genética , Integrasas/metabolismo , Riñón/anomalías , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Morfogénesis , Fenotipo , Fosfatidilinositol 3-Quinasa/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal
3.
Cell Mol Life Sci ; 74(7): 1247-1259, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27770149

RESUMEN

The sinoatrial node (SAN) is the dominant pacemaker of the heart. Abnormalities in SAN formation and function can cause sinus arrhythmia, including sick sinus syndrome and sudden death. A better understanding of genes and signaling pathways that regulate SAN development and function is essential to develop more effective treatment to sinus arrhythmia, including biological pacemakers. In this review, we briefly summarize the key processes of SAN morphogenesis during development, and focus on the transcriptional network that drives SAN development.


Asunto(s)
Cardiopatías/terapia , Marcapaso Artificial , Arritmia Sinusal/etiología , Arritmia Sinusal/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Proteínas con Homeodominio LIM/metabolismo , Marcapaso Artificial/efectos adversos , Nodo Sinoatrial/metabolismo , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo
4.
J Cell Mol Med ; 19(9): 2143-52, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26059563

RESUMEN

The voltage-gated Na(+) channel Nav 1.5 is essential for action potential (AP) formation and electrophysiological homoeostasis in the heart. The ubiquitin-proteasome system (UPS) is a major degradative system for intracellular proteins including ion channels. The ubiquitin protein ligase E3 component N-recognin (UBR) family is a part of the UPS; however, their roles in regulating cardiac Nav 1.5 channels remain elusive. Here, we found that all of the UBR members were expressed in cardiomyocytes. Individual knockdown of UBR3 or UBR6, but not of other UBR members, significantly increased Nav 1.5 protein levels in neonatal rat ventricular myocytes, and this effect was verified in HEK293T cells expressing Nav 1.5 channels. The UBR3/6-dependent regulation of Nav 1.5 channels was not transcriptionally mediated, and pharmacological inhibition of protein biosynthesis failed to counteract the increase in Nav 1.5 protein caused by UBR3/6 reduction, suggesting a degradative modulation of UBR3/6 on Nav 1.5. Furthermore, the effects of UBR3/6 knockdown on Nav 1.5 proteins were abolished under the inhibition of proteasome activity, and UBR3/6 knockdown reduced Nav 1.5 ubiquitylation. The double UBR3-UBR6 knockdown resulted in comparable increases in Nav 1.5 proteins to that observed for single knockdown of either UBR3 or UBR6. Electrophysiological recordings showed that UBR3/6 reduction-mediated increase in Nav 1.5 protein enhanced the opening of Nav 1.5 channels and thereby the amplitude of the AP. Thus, our findings indicate that UBR3/6 regulate cardiomyocyte Nav 1.5 channel protein levels via the ubiquitin-proteasome pathway. It is likely that UBR3/6 have the potential to be a therapeutic target for cardiac arrhythmias.


Asunto(s)
Miocardio/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Proteínas de Neoplasias/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Potenciales de Acción , Animales , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Miocitos Cardíacos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , ARN Interferente Pequeño/metabolismo , Ratas Sprague-Dawley
5.
Circ Res ; 113(4): 399-407, 2013 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-23743334

RESUMEN

RATIONALE: To date, there has been no specific marker of the first heart field to facilitate understanding of contributions of the first heart field to cardiac lineages. Cardiac arrhythmia is a leading cause of death, often resulting from abnormalities in the cardiac conduction system (CCS). Understanding origins and identifying markers of CCS lineages are essential steps toward modeling diseases of the CCS and for development of biological pacemakers. OBJECTIVE: To investigate HCN4 as a marker for the first heart field and for precursors of distinct components of the CCS, and to gain insight into contributions of first and second heart lineages to the CCS. METHODS AND RESULTS: HCN4CreERT2, -nuclear LacZ, and -H2BGFP mouse lines were generated. HCN4 expression was examined by means of immunostaining with HCN4 antibody and reporter gene expression. Lineage studies were performed using HCN4CreERT2, Isl1Cre, Nkx2.5Cre, and Tbx18Cre, coupled to coimmunostaining with CCS markers. Results demonstrated that, at cardiac crescent stages, HCN4 marks the first heart field, with HCN4CreERT2 allowing assessment of cell fates adopted by first heart field myocytes. Throughout embryonic development, HCN4 expression marked distinct CCS precursors at distinct stages, marking the entire CCS by late fetal stages. We also noted expression of HCN4 in distinct subsets of endothelium at specific developmental stages. CONCLUSIONS: This study provides insight into contributions of first and second heart lineages to the CCS and highlights the potential use of HCN4 in conjunction with other markers for optimization of protocols for generation and isolation of specific conduction system precursors.


Asunto(s)
Sistema de Conducción Cardíaco/citología , Sistema de Conducción Cardíaco/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Miocitos Cardíacos/metabolismo , Células Madre/metabolismo , Animales , Relojes Biológicos/genética , Biomarcadores/metabolismo , Linaje de la Célula , Femenino , Técnicas de Sustitución del Gen , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Operón Lac/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Miocitos Cardíacos/citología , Células Madre/citología
6.
J Cell Mol Med ; 18(9): 1830-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24889693

RESUMEN

Tetralogy of Fallot (TOF) is a complex congenital heart defect and the microRNAs regulation in TOF development is largely unknown. Herein, we explored the role of miRNAs in TOF. Among 75 dysregulated miRNAs identified from human heart tissues, miRNA-940 was the most down-regulated one. Interestingly, miRNA-940 was most highly expressed in normal human right ventricular out-flow tract comparing to other heart chambers. As TOF is caused by altered proliferation, migration and/or differentiation of the progenitor cells of the secondary heart field, we isolated Sca-1(+) human cardiomyocyte progenitor cells (hCMPC) for miRNA-940 function analysis. miRNA-940 reduction significantly promoted hCMPCs proliferation and inhibited hCMPCs migration. We found that JARID2 is an endogenous target regulated by miRNA-940. Functional analyses showed that JARID2 also affected hCMPCs proliferation and migration. Thus, decreased miRNA-940 affects the proliferation and migration of the progenitor cells of the secondary heart field by targeting JARID2 and potentially leads to TOF development.


Asunto(s)
Células Madre Adultas/fisiología , MicroARNs/genética , Complejo Represivo Polycomb 2/genética , Tetralogía de Fallot/metabolismo , Apoptosis , Secuencia de Bases , Sitios de Unión , Estudios de Casos y Controles , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Regulación hacia Abajo , Humanos , MicroARNs/metabolismo , Miocitos Cardíacos/fisiología , Complejo Represivo Polycomb 2/metabolismo , Interferencia de ARN , Tetralogía de Fallot/genética , Tetralogía de Fallot/patología , Transcriptoma
7.
Nature ; 454(7200): 104-8, 2008 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-18480752

RESUMEN

Understanding the origins and roles of cardiac progenitor cells is important for elucidating the pathogenesis of congenital and acquired heart diseases. Moreover, manipulation of cardiac myocyte progenitors has potential for cell-based repair strategies for various myocardial disorders. Here we report the identification in mouse of a previously unknown cardiac myocyte lineage that derives from the proepicardial organ. These progenitor cells, which express the T-box transcription factor Tbx18, migrate onto the outer cardiac surface to form the epicardium, and then make a substantial contribution to myocytes in the ventricular septum and the atrial and ventricular walls. Tbx18-expressing cardiac progenitors also give rise to cardiac fibroblasts and coronary smooth muscle cells. The pluripotency of Tbx18 proepicardial cells provides a theoretical framework for applying these progenitors to effect cardiac repair and regeneration.


Asunto(s)
Linaje de la Célula , Miocardio/citología , Miocitos Cardíacos/citología , Pericardio/citología , Pericardio/metabolismo , Células Madre/citología , Proteínas de Dominio T Box/metabolismo , Animales , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Corazón/crecimiento & desarrollo , Operón Lac/genética , Ratones , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos del Músculo Liso/metabolismo , Células Madre/metabolismo , Proteínas de Dominio T Box/genética
8.
BMC Biol ; 11: 107, 2013 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-24131868

RESUMEN

BACKGROUND: Neural crest defects lead to congenital heart disease involving outflow tract malformation. Integrin-linked-kinase (ILK) plays important roles in multiple cellular processes and embryogenesis. ILK is expressed in the neural crest, but its role in neural crest and outflow tract morphogenesis remains unknown. RESULTS: We ablated ILK specifically in the neural crest using the Wnt1-Cre transgene. ILK ablation resulted in abnormal migration and overpopulation of neural crest cells in the pharyngeal arches and outflow tract and a significant reduction in the expression of neural cell adhesion molecule (NCAM) and extracellular matrix components. ILK mutant embryos exhibited an enlarged common arterial trunk and ventricular septal defect. Reduced smooth muscle differentiation, but increased ossification and neurogenesis/innervation were observed in ILK mutant outflow tract that may partly be due to reduced transforming growth factor ß2 (TGFß2) but increased bone morphogenetic protein (BMP) signaling. Consistent with these observations, microarray analysis of fluorescence-activated cell sorting (FACS)-sorted neural crest cells revealed reduced expression of genes associated with muscle differentiation, but increased expression of genes of neurogenesis and osteogenesis. CONCLUSIONS: Our results demonstrate that ILK plays essential roles in neural crest and outflow tract development by mediating complex crosstalk between cell matrix and multiple signaling pathways. Changes in these pathways may collectively result in the unique neural crest and outflow tract phenotypes observed in ILK mutants.


Asunto(s)
Diferenciación Celular , Movimiento Celular , Cresta Neural/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Adhesión Celular , Embrión de Mamíferos , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Músculo Liso/citología , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Cresta Neural/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta2/metabolismo , Proteína Wnt1/genética
9.
Adv Sci (Weinh) ; : e2400238, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38923264

RESUMEN

The placenta links feto-maternal circulation for exchanges of nutrients, gases, and metabolic wastes between the fetus and mother, being essential for pregnancy process and maintenance. The allantois and mesodermal components of amnion, chorion, and yolk sac are derived from extraembryonic mesoderm (Ex-Mes), however, the mechanisms contributing to distinct components of the placenta and regulation the interactions between allantois and epithelium during chorioallantoic fusion and labyrinth formation remains unclear. Isl1 is expressed in progenitors of the Ex-Mes and allantois the Isl1 mut mouse line is analyzed to investigate contribution of Isl1+ Ex-Mes / allantoic progenitors to cells of the allantois and placenta. This study shows that Isl1 identifies the Ex-Mes progenitors for endothelial and vascular smooth muscle cells, and most of the mesenchymal cells of the placenta and umbilical cord. Deletion of Isl1 causes defects in allantois growth, chorioallantoic fusion, and placenta vessel morphogenesis. RNA-seq and CUT&Tag analyses revealed that Isl1 promotes allantoic endothelial, inhibits mesenchymal cell differentiation, and allantoic signals regulated by Isl1 mediating the inductive interactions between the allantois and chorion critical for chorionic epithelium differentiation, villous formation, and labyrinth angiogenesis. This study above reveals that Isl1 plays roles in regulating multiple genetic and epigenetic pathways of vascular morphogenesis, provides the insight into the mechanisms for placental formation, highlighting the necessity of Isl1 for placenta formation/pregnant maintenance.

10.
J Cell Mol Med ; 17(9): 1119-27, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23837875

RESUMEN

Sarcolemmal Na(+) /H(+) exchanger 1 (NHE1) activity is essential for the intracellular pH (pHi ) homeostasis in cardiac myocytes. Emerging evidence indicates that sarcolemmal NHE1 dysfunction was closely related to cardiomyocyte death, but it remains unclear whether defective trafficking of NHE1 plays a role in the vital cellular signalling processes. Dynamin (DNM), a large guanosine triphosphatase (GTPase), is best known for its roles in membrane trafficking events. Herein, using co-immunoprecipitation, cell surface biotinylation and confocal microscopy techniques, we investigated the potential regulation on cardiac NHE1 activity by DNM. We identified that DNM2, a cardiac isoform of DNM, directly binds to NHE1. Overexpression of a wild-type DNM2 or a dominant-negative DNM2 mutant with defective GTPase activity in adult rat ventricular myocytes (ARVMs) facilitated or retarded the internalization of sarcolemmal NHE1, whereby reducing or increasing its activity respectively. Importantly, the increased NHE1 activity associated with DNM2 deficiency led to ARVMs apoptosis, as demonstrated by cell viability, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling assay, Bcl-1/Bax expression and caspase-3 activity, which were effectively rescued by pharmacological inhibition of NHE1 with zoniporide. Thus, our results demonstrate that disruption of the DNM2-dependent retrograde trafficking of NHE1 contributes to cardiomyocyte apoptosis.


Asunto(s)
Apoptosis , Dinamina II/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Supervivencia Celular , Dinamina II/deficiencia , Células HEK293 , Ventrículos Cardíacos/citología , Humanos , Masculino , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Sarcolema/metabolismo
11.
J Mol Cell Cardiol ; 53(6): 751-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22982025

RESUMEN

Human cardiomyocyte progenitor cells (hCMPCs) are cardiac progenitor cells that are unique for their efficient differentiation into beating cardiomyocytes without requiring co-culture with neonatal cardiomyocytes. hCMPCs have shown great potential in preserving the function of infarcted mouse myocardium. MiRNA-204 has been reported to be up-regulated in differentiated hCMPCs, however, its biological significance is unclear. In this study, hCMPC proliferation, viability, apoptosis and necrosis were determined using the ELISA Kit (colorimetric BrdU detection), Cell Counting Kit-8, and Annexin V and propidium iodide staining, respectively. MiRNA-204 inhibition promoted hCMPC proliferation without affecting cell viability and the level of apoptosis and necrosis, indicating that miRNA-204 might be required for hCMPC differentiation. Quantitative reverse transcriptase-polymerase chain reactions were used to detect the expression profile of cardiac genes, including MEF2C, GATA-4, Nkx-2.5, TropT, ßMHC, and cActin. Cardiac α-actin staining was used to quantify the degree of differentiation. MiRNA-204 inhibition significantly down-regulated TropT, ßMHC, and cActin and reduced differentiation by 47.81% after 2 weeks of differentiation induction. Interestingly, miRNA-204 mimics (30 nM) did not promote hCMPC proliferation and differentiation. The bioinformatic tool GOmir identified the activating transcription factor 2 (ATF-2) as a potential target, which was confirmed by Western blot and a luciferase reporter assay. ATF-2 overexpression promoted hCMPC proliferation, further demonstrating the role played by ATF-2 as a target gene of miRNA-204. Therefore, miRNA-204 is required for hCMPC differentiation and ATF-2 is a target gene of miRNA-204 in hCMPCs. This study indicates that miRNA-204 is among the regulators that drive hCMPC proliferation and differentiation, and miRNA-204 might be used to influence cell fate.


Asunto(s)
Diferenciación Celular/genética , MicroARNs/genética , Mioblastos Cardíacos/citología , Mioblastos Cardíacos/metabolismo , Factor de Transcripción Activador 2/genética , Factor de Transcripción Activador 2/metabolismo , Animales , Apoptosis/genética , Secuencia de Bases , Supervivencia Celular/genética , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Miocardio/metabolismo , Necrosis/genética , Alineación de Secuencia
12.
Hum Mol Genet ; 19(4): 648-56, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19942620

RESUMEN

Congenital heart defects comprise the most common form of major birth defects, affecting 0.7% of all newborn infants. Jacobsen syndrome (11q-) is a rare chromosomal disorder caused by deletions in distal 11q. We have previously determined that a wide spectrum of the most common congenital heart defects occur in 11q-, including an unprecedented high frequency of hypoplastic left heart syndrome (HLHS). We identified an approximately 7 Mb 'cardiac critical region' in distal 11q that contains a putative causative gene(s) for congenital heart disease. In this study, we utilized chromosomal microarray mapping to characterize three patients with 11q- and congenital heart defects that carry interstitial deletions overlapping the 7 Mb cardiac critical region. We propose that this 1.2 Mb region of overlap harbors a gene(s) that causes at least a subset of the congenital heart defects that occur in 11q-. We demonstrate that one gene in this region, ETS-1 (a member of the ETS family of transcription factors), is expressed in the endocardium and neural crest during early mouse heart development. Gene-targeted deletion of ETS-1 in mice in a C57/B6 background causes, with high penetrance, large membranous ventricular septal defects and a bifid cardiac apex, and less frequently a non-apex-forming left ventricle (one of the hallmarks of HLHS). Our results implicate an important role for the ETS-1 transcription factor in mammalian heart development and should provide important insights into some of the most common forms of congenital heart disease.


Asunto(s)
Eliminación de Gen , Defectos del Tabique Interventricular/genética , Ventrículos Cardíacos/anomalías , Síndrome de Deleción Distal 11q de Jacobsen/genética , Proteína Proto-Oncogénica c-ets-1/genética , Animales , Deleción Cromosómica , Mapeo Cromosómico , Cromosomas Humanos Par 11/genética , Defectos del Tabique Interventricular/embriología , Defectos del Tabique Interventricular/metabolismo , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/metabolismo , Humanos , Síndrome de Deleción Distal 11q de Jacobsen/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Proto-Oncogénica c-ets-1/metabolismo
13.
Mol Cell Neurosci ; 47(3): 215-22, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21569850

RESUMEN

The LIM homeodomain transcription factor Islet1 (Isl1) is expressed in multiple organs and plays essential roles during embryogenesis. Isl1 is required for the survival and specification of spinal cord motor neurons. Due to early embryonic lethality and loss of motor neurons, the role of Isl1 in other aspects of motor neuron development remains unclear. In this study, we generated Isl1 mutant mouse lines expressing graded doses of Isl1. Our study has revealed essential roles of Isl1 in multiple aspects of motor neuron development, including motor neuron cell body localization, motor column formation and axon growth. In addition, Isl1 is required for survival of cranial ganglia neurons.


Asunto(s)
Desarrollo Embrionario/fisiología , Proteínas de Homeodominio/metabolismo , Neuronas Motoras/metabolismo , Médula Espinal/metabolismo , Animales , Axones/metabolismo , Proteínas de Homeodominio/genética , Proteínas con Homeodominio LIM , Ratones , Ratones Transgénicos , Médula Espinal/embriología , Factores de Transcripción
14.
J Cell Mol Med ; 15(12): 2712-22, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21255264

RESUMEN

Adrenergic receptor (AR)-mediated signalling is modulated by oxygen levels. Prolyl hydroxylases (PHDs) are crucial for intracellular oxygen sensing and organism survival. However, it remains to be clarified whether or how PHDs are involved in the regulation of ß(2) -adrenoceptor (ß(2) -AR) signalling. Here we show that PHD2 can modulate the rate of ß(2) -AR internalization through interactions with ß-arrestin 2. PHD2 hydroxylates ß-arrestin 2 at the proline (Pro)(176), Pro(179) and Pro(181) sites, which retards the recruitment of ß-arrestin 2 to the plasma membrane and inhibits subsequent co-internalization with ß(2) -AR into the cytosol. ß(2) -AR internalization is critical to control the temporal and spatial aspects of ß(2) -AR signalling. Identifying novel regulators of ß(2) -AR internalization will enable us to develop new strategies to manipulate receptor signalling and provide potential targets for drug development in the prevention and treatment of diseases associated with ß(2) -AR signalling dysregulation.


Asunto(s)
Arrestinas/metabolismo , Membrana Celular/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Células Cultivadas , Citosol/metabolismo , Endocitosis , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Inmunoprecipitación , Riñón/citología , Riñón/metabolismo , Fosforilación , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Procolágeno-Prolina Dioxigenasa/genética , ARN Interferente Pequeño/genética , Transducción de Señal , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Arrestina beta 2 , beta-Arrestinas
15.
Physiol Genomics ; 43(11): 655-64, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21325063

RESUMEN

The aim of this study was to investigate the microRNA (miRNA) signature in atrial fibrillation (AF) with mitral stenosis (MS). miRNA arrays were used to evaluate the expression signature of the right atrial appendages of healthy individuals (n=9), patients with MS and AF (n=9) and patients with MS without AF (n=4). The results were validated with qRT-PCR analysis. GOmir was used to predict the potential miRNA targets and to analyze their functions. DIANA-mirPath was used to incorporate the miRNAs into pathways. miRNA arrays revealed that 136 and 96 miRNAs were expressed at different levels in MS patients with AF and in MS patients without AF, respectively, compared with healthy controls. More importantly, 28 miRNAs were expressed differently in the MS patients with AF compared with the MS patients without AF; of these miRNAs, miR-1202 was the most dysregulated. The unsupervised hierarchical clustering analysis based on the 28 differently expressed miRNAs showed that the heat map of miRNA expression categorized two well-defined clusters that corresponded to MS with AF and MS without AF. The qRT-PCR results correlated well with the microarray data. Bioinformatic analysis indicated the potential miRNA targets and molecular pathways. This study shows that there is a distinct miRNA expression signature in AF with MS. The findings may be useful for the development of therapeutic interventions that are based on rational target selection in these patients.


Asunto(s)
Fibrilación Atrial/genética , MicroARNs/genética , Estenosis de la Válvula Mitral/genética , Adulto , Anciano , Fibrilación Atrial/complicaciones , Fibrilación Atrial/metabolismo , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Estenosis de la Válvula Mitral/etiología , Estenosis de la Válvula Mitral/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos
16.
J Clin Invest ; 118(12): 3870-80, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19033658

RESUMEN

The response of cardiomyocytes to biomechanical stress can determine the pathophysiology of hypertrophic cardiac disease, and targeting the pathways regulating these responses is a therapeutic goal. However, little is known about how biomechanical stress is sensed by the cardiomyocyte sarcomere to transduce intracellular hypertrophic signals or how the dysfunction of these pathways may lead to disease. Here, we found that four-and-a-half LIM domains 1 (FHL1) is part of a complex within the cardiomyocyte sarcomere that senses the biomechanical stress-induced responses important for cardiac hypertrophy. Mice lacking Fhl1 displayed a blunted hypertrophic response and a beneficial functional response to pressure overload induced by transverse aortic constriction. A link to the Galphaq (Gq) signaling pathway was also observed, as Fhl1 deficiency prevented the cardiomyopathy observed in Gq transgenic mice. Mechanistic studies demonstrated that FHL1 plays an important role in the mechanism of pathological hypertrophy by sensing biomechanical stress responses via the N2B stretch sensor domain of titin and initiating changes in the titin- and MAPK-mediated responses important for sarcomere extensibility and intracellular signaling. These studies shed light on the physiological regulation of the sarcomere in response to hypertrophic stress.


Asunto(s)
Mecanotransducción Celular , Proteínas Musculares/metabolismo , Miocitos Cardíacos/metabolismo , Sarcómeros/metabolismo , Estrés Fisiológico , Animales , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas con Dominio LIM , Sistema de Señalización de MAP Quinasas/genética , Mecanotransducción Celular/genética , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Estrés Fisiológico/genética
17.
J Transl Med ; 9: 159, 2011 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-21943159

RESUMEN

BACKGROUND: Acute pulmonary embolism (APE) remains a diagnostic challenge due to a variable clinical presentation and the lack of a reliable screening tool. MicroRNAs (miRNAs) regulate gene expression in a wide range of pathophysiologic processes. Circulating miRNAs are emerging biomarkers in heart failure, type 2 diabetes and other disease states; however, using plasma miRNAs as biomarkers for the diagnosis of APE is still unknown. METHODS: Thirty-two APE patients, 32 healthy controls, and 22 non-APE patients (reported dyspnea, chest pain, or cough) were enrolled in this study. The TaqMan miRNA microarray was used to identify dysregulated miRNAs in the plasma of APE patients. The TaqMan-based miRNA quantitative real-time reverse transcription polymerase chain reactions were used to validate the dysregulated miRNAs. The receiver-operator characteristic (ROC) curve analysis was conducted to evaluate the diagnostic accuracy of the miRNA identified as the candidate biomarker. RESULTS: Plasma miRNA-134 (miR-134) level was significantly higher in the APE patients than in the healthy controls or non-APE patients. The ROC curve showed that plasma miR-134 was a specific diagnostic predictor of APE with an area under the curve of 0.833 (95% confidence interval, 0.737 to 0.929; P < 0.001). CONCLUSIONS: Our findings indicated that plasma miR-134 could be an important biomarker for the diagnosis of APE. Because of this finding, large-scale investigations are urgently needed to pave the way from basic research to clinical utilization.


Asunto(s)
MicroARNs/sangre , Embolia Pulmonar/sangre , Embolia Pulmonar/diagnóstico , Enfermedad Aguda , Biomarcadores/sangre , Estudios de Casos y Controles , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Curva ROC , Reproducibilidad de los Resultados , Factores de Riesgo
18.
Circulation ; 120(7): 568-76, 2009 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-19652092

RESUMEN

BACKGROUND: PINCH proteins are 5 LIM domain-only adaptor proteins that function as key components of the integrin signaling pathway and play crucial roles in multiple cellular processes. Two PINCH proteins, PINCH1 and PINCH2, have been described in mammals and share high homology. Both PINCH1 and PINCH2 are ubiquitously expressed in most tissues and organs, including myocardium. Cardiac-specific PINCH1 knockout or global PINCH2 knockout mice exhibit no basal cardiac phenotype, which may reflect a redundant role for these 2 PINCH proteins in myocardium. A potential role for PINCH proteins in myocardium remains unknown. METHODS AND RESULTS: To define the role of PINCH in myocardium, we generated mice that were doubly homozygous null for PINCH1 and PINCH2 in myocardium. Resulting mutants were viable at birth but developed dilated cardiomyopathy and died of heart failure within 4 weeks. Mutant hearts exhibited disruptions of intercalated disks and costameres accompanied by fibrosis. Furthermore, multiple cell adhesion proteins exhibited reduced expression and were mislocalized. Mutant cardiomyocytes were significantly smaller and irregular in size. In addition, we observed that the absence of either PINCH1 or PINCH2 in myocardium leads to exacerbated cardiac injury and deterioration in cardiac function after myocardial infarction. CONCLUSIONS: These results demonstrate essential roles for PINCHs in myocardial growth, maturation, remodeling, and function and highlight the importance of studying the role of PINCHs in human cardiac injury and cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Insuficiencia Cardíaca/etiología , Miocardio/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/mortalidad , Adhesión Celular/fisiología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/mortalidad , Integrinas/metabolismo , Proteínas con Dominio LIM , Proteínas de la Membrana , Ratones , Ratones Noqueados , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Miocardio/ultraestructura , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología
19.
Biochem Biophys Res Commun ; 401(2): 231-7, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20849813

RESUMEN

Prolyl hydroxylases (PHDs) are dioxygenases that use oxygen as a co-substrate to hydroxylate proline residues. Three PHD isoforms (PHD1, PHD2 and PHD3) have been identified in mammalian cells. PHD3 expression is upregulated in some cardiac diseases such as cardiomyopathy, myocardial ischemia-reperfusion injury and congestive heart failure, all of which are associated with apoptosis. However, the role of PHDs in cardiomyocyte apoptosis remains unknown. Here, we have found that exposure of embryonic rat heart-derived H9c2 cells to doxorubicin (DOX) induced cell apoptosis as evaluated by caspase-3/7 activity, mitochondrial membrane potential (Δψm) and cell viability, and that this apoptosis was linked to PHD3 upregulation. PHD inhibition or PHD3 silencing substantially ameliorated DOX-induced apoptosis, but PHD1 or PHD2 knockdown did not significantly influence apoptosis. Furthermore, immunoprecipitation experiments showed that PHD3 upregulation reduced the formation of the Bax-Bcl-2 complex, inhibiting the anti-apoptotic effect of Bcl-2. Thus, PHD3 upregulation may be partially responsible for DOX-induced cardiomyocyte apoptosis via its interaction with Bcl-2. Inhibition of PHD3 is likely to be cardioprotective against apoptosis in some heart disorders.


Asunto(s)
Apoptosis , Miocitos Cardíacos/fisiología , Procolágeno-Prolina Dioxigenasa/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Línea Celular , Doxorrubicina/farmacología , Humanos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/genética , Ratas
20.
Dev Cell ; 5(6): 877-89, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14667410

RESUMEN

Hearts of mice lacking Isl1, a LIM homeodomain transcription factor, are completely missing the outflow tract, right ventricle, and much of the atria. isl1 expression and lineage tracing of isl1-expressing progenitors demonstrate that Isl1 is a marker for a distinct population of undifferentiated cardiac progenitors that give rise to the cardiac segments missing in isl1 mutants. Isl1 function is required for these progenitors to contribute to the heart. In isl1 mutants, isl1-expressing progenitors are progressively reduced in number, and FGF and BMP growth factors are downregulated. Our studies define two sets of cardiogenic precursors, one of which expresses and requires Isl1 and the other of which does not. Our results have implications for the development of specific cardiac lineages, left-right asymmetry, cardiac evolution, and isolation of cardiac progenitor cells.


Asunto(s)
Cardiopatías Congénitas/genética , Corazón/embriología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Miocardio/citología , Proteínas del Tejido Nervioso , Células Madre/fisiología , Animales , Diferenciación Celular/fisiología , División Celular/fisiología , Linaje de la Célula/fisiología , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Endodermo/citología , Femenino , Regulación del Desarrollo de la Expresión Génica , Homocigoto , Proteínas con Homeodominio LIM , Mesodermo/citología , Ratones , Ratones Noqueados , Faringe/citología , Faringe/embriología , Embarazo , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA