Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Genesis ; 49(11): 841-50, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21504044

RESUMO

The Bmp2 3'untranslated region (UTR) sequence bears a sequence conserved between mammals and fishes that can post-transcriptionally activate or repress protein synthesis. We developed a map of embryonic cells in the mouse where this potent Bmp2 regulatory sequence functions by using a lacZ reporter transgene with a 3'UTR bearing two loxP sites flanking the ultra-conserved sequence. Cre-recombinase-mediated deletion of the ultra-conserved sequence caused strong ectopic expression in proepicardium, epicardium and epicardium-derived cells (EPDC) and in tissues with known epicardial contributions (coronary vessels and valves). Transient transfections of reporters in the epicardial/mesothelial cell (EMC) line confirmed this repression. Ectopic expression of the recombined transgene also occurred in the aorta, outlet septum, posterior cardiac plexus, cardiac and extracardiac nerves and neural ganglia. Bmp2 is dynamically regulated in the developing heart. 3'UTR-mediated mechanisms that restrain BMP2 synthesis may be relevant to congenital heart and vasculature malformations and to adult diseases involving aberrant BMP2 synthesis.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Coração/fisiologia , Pericárdio/metabolismo , Regiões 3' não Traduzidas , Animais , Proteína Morfogenética Óssea 2/genética , Linhagem Celular , Sequência Conservada , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Coração/embriologia , Coração/inervação , Imuno-Histoquímica , Integrases/metabolismo , Óperon Lac , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/genética , Camundongos Transgênicos/metabolismo , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/metabolismo , Pericárdio/citologia , Pericárdio/embriologia , Pericárdio/fisiologia , Processamento de Proteína Pós-Traducional , Ratos , Sequências Reguladoras de Ácido Nucleico , Deleção de Sequência , Transcrição Gênica , Transfecção , Transgenes
2.
J Cell Biochem ; 112(2): 666-74, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21268088

RESUMO

BMP2 is a morphogen that controls mesenchymal cell differentiation and behavior. For example, BMP2 concentration controls the differentiation of mesenchymal precursors into myocytes, adipocytes, chondrocytes, and osteoblasts. Sequences within the 3'untranslated region (UTR) of the Bmp2 mRNA mediate a post-transcriptional block of protein synthesis. Interaction of cell and developmental stage-specific trans-regulatory factors with the 3'UTR is a nimble and versatile mechanism for modulating this potent morphogen in different cell types. We show here, that an ultra-conserved sequence in the 3'UTR functions independently of promoter, coding region, and 3'UTR context in primary and immortalized tissue culture cells and in transgenic mice. Our findings indicate that the ultra-conserved sequence is an autonomously functioning post-transcriptional element that may be used to modulate the level of BMP2 and other proteins while retaining tissue specific regulatory elements.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Células-Tronco Mesenquimais/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Regiões 3' não Traduzidas/genética , Animais , Aorta/citologia , Proteína Morfogenética Óssea 2/genética , Linhagem Celular , Células Cultivadas , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Sequências Reguladoras de Ácido Nucleico/fisiologia
3.
Circ Res ; 104(7): 887-95, 2009 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-19246680

RESUMO

H11 kinase/Hsp22 (H11K) is a chaperone promoting cardiac cell growth and survival through the activation of Akt, a downstream effector of phosphatidylinositol 3-kinase (PI3K). In this study, we tested whether H11K-induced activation of the PI3K/Akt pathway is mediated by the bone morphogenetic protein (BMP) signaling, both in a transgenic mouse model with cardiac-specific overexpression of H11K and in isolated cardiac myocytes. Microarrays in hearts from transgenic compared to wild-type mice showed an upregulation of the BMP receptors Alk3 and BMPR-II, and of their ligand BMP4 (P<0.01 versus wild type). Activation of the BMP pathway in transgenic mice was confirmed by increased phosphorylation of the "canonical" BMP effectors Smad 1/5/8 (P<0.01 versus wild type). In isolated myocytes, adenovirus-mediated overexpression of H11K was accompanied by a significant (P<0.01) increase in PI3K activity, phospho-Akt, Smad 1/5/8 phosphorylation and [(3)H]phenylalanine incorporation, and by a 70% reduction in H(2)O(2)-mediated apoptosis. All these effects were abolished by the BMP antagonist noggin. In presence of BMP4, Smad 1/5/8 phosphorylation was enhanced by 5-fold on H11K overexpression but decreased by 3-fold on H11K knockdown (P<0.01 versus control), showing that H11K potentiates the BMP signaling. In pull-down experiments, H11K increased both the association of Alk3 and BMPR-II together, and their interaction with the transforming growth factor-beta-activated kinase (TAK)1, a "noncanonical" mediator of the BMP receptor signaling. TAK1 inhibition prevented H11K-mediated activation of Akt. Therefore, potentiation of the BMP receptor by H11K promotes an activation of the PI3K/Akt pathway mediated by TAK1, which dictates the physiological effects of H11K on cardiac cell growth and survival.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Proliferação de Células , Proteínas de Choque Térmico/metabolismo , Miócitos Cardíacos/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Animais , Animais Recém-Nascidos , Apoptose , Proteína Morfogenética Óssea 4/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Proteínas de Transporte/metabolismo , Sobrevivência Celular , Células Cultivadas , Proteínas de Choque Térmico/genética , Humanos , MAP Quinase Quinase Quinases/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares , Miócitos Cardíacos/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Proteínas Smad Reguladas por Receptor/metabolismo , Transdução Genética
4.
Am J Physiol Heart Circ Physiol ; 297(2): H785-91, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19465551

RESUMO

The goal of this investigation was to determine the distribution of myocardial apoptosis in myocytes and nonmyocytes in primates and patients with heart failure (HF). Almost all clinical cardiologists and cardiovascular investigators believe that myocyte apoptosis is considered to be a cardinal sign of HF and a major factor in its pathogenesis. However, with the knowledge that 75% of the number of cells in the heart are nonmyocytes, it is important to determine whether the apoptosis in HF is occurring in myocytes or in nonmyocytes. We studied both a nonhuman primate model of chronic HF, induced by rapid pacing 2-6 mo after myocardial infarction (MI), and biopsies from patients with ischemic cardiomyopathy. Dual labeling with a cardiac muscle marker was used to discriminate apoptosis in myocytes versus nonmyocytes. Left ventricular ejection fraction decreased following MI (from 78% to 60%) and further with HF (35%, P < 0.05). As expected, total apoptosis was increased in the myocardium following recovery from MI (0.62 cells/mm(2)) and increased further with the development of HF (1.91 cells/mm(2)). Surprisingly, the majority of apoptotic cells in MI and MI + HF, and in both the adjacent and remote areas, were nonmyocytes. This was also observed in myocardial biopsies from patients with ischemic cardiomyopathy. We found that macrophages contributed the largest fraction of apoptotic nonmyocytes (41% vs. 18% neutrophils, 16% fibroblast, and 25% endothelial and other cells). Although HF in the failing human and monkey heart is characterized by significant apoptosis, in contrast to current concepts, the apoptosis in nonmyocytes was eight- to ninefold greater than in myocytes.


Assuntos
Apoptose/fisiologia , Cardiomiopatias/patologia , Insuficiência Cardíaca/patologia , Infarto do Miocárdio/patologia , Miocárdio/patologia , Animais , Biópsia , Caspase 3/metabolismo , Modelos Animais de Doenças , Fibroblastos/patologia , Humanos , Marcação In Situ das Extremidades Cortadas , Macaca fascicularis , Macrófagos/patologia , Masculino , Isquemia Miocárdica/patologia , Miócitos Cardíacos/patologia , Neutrófilos/patologia , Marca-Passo Artificial
5.
Circulation ; 116(22): 2535-43, 2007 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-17998461

RESUMO

BACKGROUND: The atrioventricular (AV) node is essential for the sequential excitation and optimized contraction of the adult multichambered heart; however, relatively little is known about its formation from the embryonic AV canal. A recent study demonstrated that signaling by Alk3, the type 1a receptor for bone morphogenetic proteins, in the myocardium of the AV canal was required for the development of both the AV valves and annulus fibrosus. To test the hypothesis that bone morphogenetic protein signaling also plays a role in AV node formation, we investigated conduction system function and AV node morphology in adult mice with conditional deletion of Alk3 in the AV canal. METHODS AND RESULTS: High-resolution optical mapping with correlative histological analysis of 28 mutant hearts revealed 4 basic phenotypic classes based on electrical activation patterns and volume-conducted ECGs. The frequency of AV node conduction and morphological abnormalities increased from no detectable anomalies (class I) to severe defects (class IV), which included the presence of bypass tracts, abnormal ventricular activation patterns, fibrosis of the AV node, and twin AV nodes. CONCLUSIONS: The present findings demonstrate that bone morphogenetic protein signaling is required in the myocardium of the AV canal for proper AV junction development, including the AV node.


Assuntos
Nó Atrioventricular/fisiopatologia , Mapeamento Potencial de Superfície Corporal , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Sistema de Condução Cardíaco/fisiopatologia , Animais , Nó Atrioventricular/crescimento & desenvolvimento , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/fisiologia , Genótipo , Bloqueio Cardíaco , Camundongos , Camundongos Mutantes , Miocárdio/patologia
6.
Circ Res ; 98(2): 280-8, 2006 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-16373598

RESUMO

Ischemic preconditioning confers powerful protection against myocardial infarction through pre-emptive activation of survival signaling pathways, but it remains difficult to apply to patients with ischemic heart disease, and its effects are transient. Promoting a sustained activation of preconditioning mechanisms in vivo would represent a novel approach of cardioprotection. We tested the role of the protein H11 kinase (H11K), which accumulates by 4- to 6-fold in myocardium of patients with chronic ischemic heart disease and in experimental models of ischemia. This increased expression was quantitatively reproduced in cardiac myocytes using a transgenic (TG) mouse model. After 45 minutes of coronary artery occlusion and reperfusion, hearts from TG mice showed an 82+/-5% reduction in infarct size compared with wild-type (WT), which was similar to the 84+/-4% reduction of infarct size observed in WT after a protocol of ischemic preconditioning. Hearts from TG mice showed significant activation of survival kinases participating in preconditioning, including Akt and the 5'AMP-activated protein kinase (AMPK). H11K directly binds to both Akt and AMPK and promotes their nuclear translocation and their association in a multiprotein complex, which results in a stimulation of survival mechanisms in cytosol and nucleus, including inhibition of proapoptotic effectors (glycogen synthase kinase-3beta, Bad, and Foxo), activation of antiapoptotic effectors (protein kinase Cepsilon, endothelial and inducible NO synthase isoforms, and heat shock protein 70), increased expression of the hypoxia-inducible factor-1alpha, and genomic switch to glucose utilization. Therefore, activation of survival pathways by H11K preemptively triggers the antiapoptotic and metabolic response to ischemia and is sufficient to confer cardioprotection in vivo equally potent to preconditioning.


Assuntos
Proteínas de Choque Térmico HSP20/fisiologia , Precondicionamento Isquêmico Miocárdico , Proteínas Musculares/fisiologia , Infarto do Miocárdio/prevenção & controle , Proteínas Quinases Ativadas por AMP , Animais , Apoptose , Sobrevivência Celular , Células Cultivadas , Citoproteção , Proteínas de Choque Térmico , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares , Complexos Multienzimáticos/metabolismo , Miócitos Cardíacos/citologia , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
7.
Circ Res ; 97(3): 219-26, 2005 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-16037571

RESUMO

Endocardial cushions are precursors of mature atrioventricular (AV) valves. Their formation is induced by signaling molecules originating from the AV myocardium, including bone morphogenetic proteins (BMPs). Here, we hypothesized that BMP signaling plays an important role in the AV myocardium during the maturation of AV valves from the cushions. To test our hypothesis, we used a unique Cre/lox system to target the deletion of a floxed Alk3 allele, the type IA receptor for BMPs, to cardiac myocytes of the AV canal (AVC). Lineage analysis indicated that cardiac myocytes of the AVC contributed to the tricuspid mural and posterior leaflets, the mitral septal leaflet, and the atrial border of the annulus fibrosus. When Alk3 was deleted in these cells, defects were seen in the same leaflets, ie, the tricuspid mural leaflet and mitral septal leaflet were longer, the tricuspid posterior leaflet was displaced and adherent to the ventricular wall, and the annulus fibrosus was disrupted resulting in ventricular preexcitation. The defects seen in mice with AVC-targeted deletion of Alk3 provide strong support for a role of Alk3 in human congenital heart diseases, such as Ebstein's anomaly. In conclusion, our mouse model demonstrated critical roles for Alk3 signaling in the AV myocardium during the development of AV valves and the annulus fibrosus.


Assuntos
Valvas Cardíacas/embriologia , Coração/embriologia , Proteínas Serina-Treonina Quinases/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Proteínas de Ligação a DNA/genética , Fator de Transcrição GATA6 , Valvas Cardíacas/anormalidades , Integrases/genética , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/fisiologia , Transdução de Sinais , Fatores de Transcrição/genética
8.
Cytokine Growth Factor Rev ; 14(1): 1-4, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12485614

RESUMO

Heart muscle cell specification (cardiac myogenesis) and creating the four-chambered heart (cardiac morphogenesis) are subject to regulation, in certain model organisms, by bone morphogenetic proteins and their receptors. Extrapolation to mammals from organisms that develop outside the mother (flies, fish, frogs, and avians) has been confounded by very early lethality-at gastrulation-of many null alleles needed to prove cause-effect relations in this pathway. Here, we describe the use of lineage- or compartment-restricted null alleles as well as hypomorphic alleles, which circumvent these limitations and pinpoint novel essential functions for the bone morphogenetic protein cascade in mammalian cardiac development.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Coração/crescimento & desenvolvimento , Transdução de Sinais/fisiologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas , Proteínas Morfogenéticas Ósseas/genética , Marcação de Genes , Morfogênese , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento/metabolismo
9.
Circ Res ; 91(11): 1007-14, 2002 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-12456486

RESUMO

By subtractive hybridization, we found a significant increase in H11 kinase transcript in large mammalian models of both ischemia/reperfusion (stunning) and chronic pressure overload with hypertrophy. Because this gene has not been characterized in the heart, the goal of the present study was to determine the function of H11 kinase in cardiac tissue, both in vitro and in vivo. In isolated neonatal rat cardiac myocytes, adenoviral-mediated overexpression of H11 kinase resulted in a 37% increase in protein/DNA ratio, reflecting hypertrophy. A cardiac-specific transgene driven by the alphaMHC-promoter was generated, which resulted in an average 7-fold increase in H11 kinase protein expression. Transgenic hearts were characterized by a 30% increase of the heart weight/body weight ratio, by the reexpression of a fetal gene program, and by concentric hypertrophy with preserved contractile function at echocardiography. This phenotype was accompanied by a dose-dependent activation of Akt/PKB and p70(S6) kinase, whereas the MAP kinase pathway was unaffected. Thus, H11 kinase represents a novel mediator of cardiac cell growth and hypertrophy.


Assuntos
Cardiomegalia/fisiopatologia , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Cardiomegalia/etiologia , Cardiomegalia/patologia , Divisão Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Cães , Feminino , Genes Reporter , Proteínas de Choque Térmico , Hipertrofia Ventricular Esquerda/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares , Miocárdio/citologia , Miocárdio/metabolismo , Miocárdio/patologia , Hibridização de Ácido Nucleico/métodos , Especificidade de Órgãos , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/fisiologia , Transdução Genética , Regulação para Cima
10.
Circ Res ; 95(4): 433-40, 2004 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-15242971

RESUMO

Hibernating myocardium refers to chronically dysfunctional myocardium in patients with coronary artery disease in which cardiac viability is maintained and whose function improves after coronary revascularization. It is our hypothesis that long-term adaptive genomic mechanisms subtend the survival capacity of this ischemic myocardium. Therefore, the goal of this study was to determine whether chronic repetitive ischemia elicits a gene program of survival protecting hibernating myocardium against cell death. Accordingly, we measured the expression of survival genes in hibernating myocardium, both in patients surgically treated for hibernation and in a chronic swine model of repetitive ischemia reproducing the features of hibernation. Human hibernating myocardium was characterized by an upregulation of genes and corresponding proteins involved in anti-apoptosis (IAP), growth (VEGF, H11 kinase), and cytoprotection (HSP70, HIF-1alpha, GLUT1). In the swine model, the same genes and proteins were upregulated after repetitive ischemia, which was accompanied by a concomitant decrease in myocyte apoptosis. These changes characterize viable tissue, because they were not found in irreversibly injured myocardium. Our report demonstrates a novel mechanism by which the activation of an endogenous gene program of cell survival underlies the sustained viability of the hibernating heart. Potentially, promoting such a program offers a novel opportunity to salvage postmitotic tissues in conditions of ischemia.


Assuntos
Regulação da Expressão Gênica , Miocárdio Atordoado/genética , Miócitos Cardíacos/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Apoptose/genética , Sobrevivência Celular/genética , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Feminino , Perfilação da Expressão Gênica , Transportador de Glucose Tipo 1 , Proteínas de Choque Térmico HSP70/biossíntese , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Proteínas Inibidoras de Apoptose , Imagem Cinética por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Modelos Animais , Chaperonas Moleculares , Proteínas de Transporte de Monossacarídeos/biossíntese , Proteínas de Transporte de Monossacarídeos/genética , Isquemia Miocárdica/genética , Miocárdio Atordoado/diagnóstico por imagem , Miócitos Cardíacos/citologia , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Tomografia por Emissão de Pósitrons , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Proteínas/genética , Proteínas/metabolismo , RNA Mensageiro/biossíntese , Sus scrofa , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
11.
Circulation ; 108(23): 2926-33, 2003 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-14623810

RESUMO

BACKGROUND: Although beta-adrenergic receptor (AR) blockade therapy is beneficial in the treatment of heart failure, little is known regarding the transcriptional mechanisms underlying this salutary action. METHODS AND RESULTS: In the present study, we screened mice overexpressing Gsalpha, beta1AR, beta2AR, or protein kinase A to test if a common genomic pathway exists in different models with enhanced beta-adrenergic signaling. In mice overexpressing Gsalpha, differentially expressed genes were identified by mRNA profiling. In addition to well-known markers of cardiac hypertrophy (atrial natriuretic factor, CARP, and beta-myosin heavy chain), uncoupling protein 2 (UCP2), a protein involved in the control of mitochondrial membrane potential, and four-and-a-half LIM domain protein-1 (FHL1), a member of the LIM protein family, were predicted to be upregulated. Upregulation of these genes was confirmed by quantitative reverse transcriptase-polymerase chain reaction at all time points tested during the development of cardiomyopathy in mice overexpressing Gsalpha. In mice overexpressing beta1AR, beta2AR, or protein kinase A, increased UCP2 and FHL1 expression was also observed at the onset of cardiomyopathy. BetaAR blockade treatment reversed the cardiomyopathy and suppressed the increased expression of UCP2 and FHL1 in mice overexpressing Gsalpha. CONCLUSIONS: UCP2 and FHL1 are important candidate genes that correlate with the development of betaAR-induced cardiomyopathy in different mouse models with enhanced betaAR signaling. In addition to preserving cardiac function, betaAR blockade treatment also prevents the genomic regulation that correlates with the onset of heart failure.


Assuntos
Antagonistas Adrenérgicos beta/uso terapêutico , Cardiomiopatias/genética , Epinefrina/fisiologia , Regulação da Expressão Gênica , Insuficiência Cardíaca/prevenção & controle , Receptores Adrenérgicos beta/fisiologia , Antagonistas Adrenérgicos beta/farmacologia , Animais , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/etiologia , Proteínas Quinases Dependentes de AMP Cíclico/biossíntese , Proteínas Quinases Dependentes de AMP Cíclico/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/biossíntese , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Insuficiência Cardíaca/genética , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Canais Iônicos , Proteínas de Membrana Transportadoras/biossíntese , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Transgênicos , Proteínas Mitocondriais/biossíntese , Proteínas Mitocondriais/genética , Modelos Animais , Receptores Adrenérgicos beta/biossíntese , Receptores Adrenérgicos beta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Proteína Desacopladora 2
12.
Cardiovasc Res ; 58(1): 126-35, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12667953

RESUMO

BACKGROUND: Previously, we showed by subtractive hybridization in a swine model of ischemia/reperfusion that an upregulation of genes participating in mechanisms of cell survival is a potential genomic mechanism to tilt the balance from necrosis to functional reversibility. METHODS AND RESULTS: We present here the full-length sequencing and characterization of a novel gene that was found in this subtraction, encoding a cardiac-specific DnaJ-like co-chaperone that we call Pig DnaJ-like protein A1 (pDJA1). The expression of pDJA1 was found to be restricted to the heart, as opposed to skeletal muscle, liver, lung, kidney, aorta, stomach and spleen. Expression of pDJA1 is restricted to cardiac myocytes, as determined by in situ hybridization. The transcript is expressed more in the left ventricle than in the other cardiac chambers. Remarkably, expression of pDJA1 follows a transmural gradient in the left ventricle, with the highest level of expression in the subendocardium. Expression of pDJA1 slightly increased during an episode of ischemia, but increased by 4-fold during the following period of reperfusion. Adenovirus-mediated transduction of pDJA1 in isolated rat neonatal cardiac myocytes decreased by 65% the rate of apoptosis induced by staurosporine. CONCLUSION: Therefore, pDJA1 is a novel heart-specific, ventricle-enriched cardioprotective co-chaperone, which participates in the program of cell survival that limits irreversible damage in post-ischemic myocardium.


Assuntos
Proteínas de Choque Térmico/análise , Isquemia Miocárdica/metabolismo , Miocárdio/química , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting/métodos , Células Cultivadas , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Proteínas de Choque Térmico HSP40 , Proteínas de Choque Térmico/genética , Hibridização In Situ/métodos , Dados de Sequência Molecular , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Suínos , Distribuição Tecidual , Transdução Genética/métodos
13.
J Long Term Eff Med Implants ; 25(1-2): 17-26, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25955004

RESUMO

Heart valves are complex structures composed of organized layers of extracellular matrix, and interstitial and overlying endothelial cells. In this article, we present the specific localization of a population of melanocytes within the murine heart valves at ages important for their post-natal development. In all stages analyzed in our study, melanocytes were found in high numbers populating the atrial aspect of the tricuspid and mitral leaflets. The pulmonary valve did not present melanocytes. To characterize a putative role for the valve melanocytes, the dynamic nanomechanical properties of tricuspid leaftets containing large numbers or no melanocytes were measured. The stiffness coefficient of hyperpigmented leaflets was higher (11.5 GPa) than the ones from wild-type (7.5 GPa) and hypopigmented (5.5 GPa) leaflets. These results suggest that melanocytes may contribute to the mechanical properties of the heart valves. The arrangement of extracellular matrix molecules such as Collagen I and Versican B is responsible for the mechanical characteristics of the leaflets. Melanocytes were found to reside primarily in areas of Versican B expression. The patterns of expression of Collagen I and Versican B were not, however, disrupted in hyper or hypopigmented leaflets. Melanocytes may affect other extracellular matrix molecules to alter the valves' microenvironment.


Assuntos
Matriz Extracelular/metabolismo , Doenças das Valvas Cardíacas/patologia , Valvas Cardíacas/fisiopatologia , Melanócitos/patologia , Animais , Fenômenos Biomecânicos , Células Cultivadas , Modelos Animais de Doenças , Doenças das Valvas Cardíacas/fisiopatologia , Valvas Cardíacas/metabolismo , Valvas Cardíacas/patologia , Melanócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
14.
Anat Rec A Discov Mol Cell Evol Biol ; 280(2): 1022-6, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15368346

RESUMO

This review summarizes the recent advances in understanding the development and function of the cardiac conduction system using genetically modified mice. Null mice for different cardiac connexins confirmed their suspected roles in cardiac conduction and, in addition, unraveled unexpected roles in cardiac morphogenesis. Genetically modified mice with LacZ-labeled conduction system cells are indispensable tools to the further understanding of the mechanisms governing the development of this system in the mammalian heart. Mouse models also addressed the role and contribution of specific signaling molecules, such as PRKAG2, Nkx2.5, ALK3, and Tbx5, in the development of the cardiac conduction system and the pathogenesis of cardiac dysfunction in humans.


Assuntos
Arritmias Cardíacas/patologia , Conexinas/fisiologia , Bloqueio Cardíaco/metabolismo , Sistema de Condução Cardíaco , Miócitos Cardíacos/metabolismo , Animais , Arritmias Cardíacas/metabolismo , Conexinas/genética , Morte Súbita Cardíaca/etiologia , Regulação da Expressão Gênica no Desenvolvimento , Sistema de Condução Cardíaco/embriologia , Sistema de Condução Cardíaco/fisiologia , Humanos , Modelos Animais
15.
Int J Dev Biol ; 57(11-12): 853-63, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24623077

RESUMO

Trabeculation and the formation of the leaflets of the mitral and tricuspid valves both involve remodeling of the embryonic myocardium. The nature and possible connection of these myocardial remodeling processes, however, are unclear. Therefore, we examined the morphogenesis of the early ventricular and atrioventricular (AV) myocardium and report for the first time that the formation of the early trabeculae and the positioning of the valve primordia (endocardial cushions) into the ventricular lumen are part of one continuous myocardial remodeling process, which involves the dissociation of the myocardial layers. For the endocardial cushions, this process results in delamination from the AV myocardium. The AV myocardium that will harbor the right lateral cushion is the exception and becomes positioned in the ventricular lumen by folding of the right ventricle. As a consequence, remodeling of the left and right AV myocardium occurs differently with implications for the formation of the mural leaflets and annulus fibrosis. At both the right and left side, the valvular myocardium harbors a distinct molecular phenotype and its removal from the cardiac leaflets involves a second wave of delamination. Interestingly, in the TGFß2-KO mouse, which is a known model for cushion and valve defects, remodeling of the early myocardium is disturbed as indicated by defective trabeculae formation, persistence of valvular myocardium, disturbed myocardial phenotypes and differential defects at left and right side of the AV canal. Based on these results we propose a new model clarifying early trabeculae formation and AV valve formation and provide new inroads for an enhanced understanding of congenital heart defects.


Assuntos
Valvas Cardíacas/embriologia , Valvas Cardíacas/patologia , Miocárdio/patologia , Proteínas com Domínio T/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Animais , Apoptose , Endocárdio/embriologia , Coração/embriologia , Átrios do Coração/patologia , Ventrículos do Coração/patologia , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Fatores de Tempo , Remodelação Ventricular/fisiologia
17.
Ann Biomed Eng ; 38(3): 674-82, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20099034

RESUMO

Valvular heart disease is a leading cause of morbidity and mortality in adults but little is known about the underlying etiology. A better understanding of the genetic and hemodynamic mechanisms involved in growth and remodeling of heart valves during physiological and pathological conditions is needed for a better understanding of valvular heart disease. Here, we report the design of a miniature tissue culture system (MTCS) that allows the culture of mitral valves from perinatal to adult mice. The design of the MTCS is novel in that fine positioning and cannulation can be conducted with hearts of different sizes (perinatal to adult). Perfusion of the heart and hence, culture of the mitral valve in its natural position, occurs in a hydraulically sealed culture bath environment. Using the MTCS, we successfully cultured the mitral valve of adult mouse hearts for 3 days. Histological analysis indicated that the cultured valves remained viable and their extracellular matrix organization was similar to age-matched native valves. Gene expression could also be modified in cultured valves by perfusion with medium containing beta-galactosidase-expressing adenovirus. Thus, the MTCS is a new tool to study the genetic and hemodynamic mechanisms underlying the three-dimensional organization of the heart valves, which could provide insights in the pathology of valvular heart disease and be used in animal models for the development of tissue-engineered heart valves.


Assuntos
Bioprótese/veterinária , Próteses Valvulares Cardíacas/veterinária , Miócitos Cardíacos/fisiologia , Técnicas de Cultura de Órgãos/instrumentação , Perfusão/instrumentação , Perfusão/veterinária , Animais , Células Cultivadas , Desenho de Equipamento , Análise de Falha de Equipamento , Camundongos , Miniaturização , Miócitos Cardíacos/citologia , Técnicas de Cultura de Órgãos/métodos
18.
J Am Coll Cardiol ; 56(9): 721-34, 2010 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-20723802

RESUMO

OBJECTIVES: The goal of this study was to guide bone marrow-derived human mesenchymal stem cells (hMSCs) into a cardiac progenitor phenotype and assess therapeutic benefit in chronic myocardial infarction. BACKGROUND: Adult stem cells, delivered in their naïve state, demonstrate a limited benefit in patients with ischemic heart disease. Pre-emptive lineage pre-specification may optimize therapeutic outcome. METHODS: hMSC were harvested from a coronary artery disease patient cohort. A recombinant cocktail consisting of transforming growth factor-beta(1), bone morphogenetic protein-4, activin A, retinoic acid, insulin-like growth factor-1, fibroblast growth factor-2, alpha-thrombin, and interleukin-6 was formulated to engage hMSC into cardiopoiesis. Derived hMSC were injected into the myocardium of a nude infarcted murine model and followed over 1 year for functional and structural end points. RESULTS: Although the majority of patient-derived hMSC in their native state demonstrated limited effect on ejection fraction, stem cells from rare individuals harbored a spontaneous capacity to improve contractile performance. This reparative cytotype was characterized by high expression of homeobox transcription factor Nkx-2.5, T-box transcription factor TBX5, helix-loop-helix transcription factor MESP1, and myocyte enhancer factor MEF2C, markers of cardiopoiesis. Recombinant cardiogenic cocktail guidance secured the cardiopoietic phenotype across the patient cohort. Compared with unguided counterparts, cardiopoietic hMSC delivered into infarcted myocardium achieved superior functional and structural benefit without adverse side effects. Engraftment into murine hearts was associated with increased human-specific nuclear, sarcomeric, and gap junction content along with induction of myocardial cell cycle activity. CONCLUSIONS: Guided cardiopoiesis thus enhances the therapeutic benefit of bone marrow-derived hMSC in chronic ischemic cardiomyopathy.


Assuntos
Transplante de Medula Óssea , Transplante de Células-Tronco Mesenquimais , Infarto do Miocárdio/cirurgia , Miócitos Cardíacos/citologia , Fatores de Transcrição/fisiologia , Animais , Fatores Biológicos/farmacologia , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Linhagem da Célula , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/fisiologia , Humanos , Proteínas de Domínio MADS/fisiologia , Fatores de Transcrição MEF2 , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , Mimetismo Molecular , Infarto do Miocárdio/genética , Miócitos Cardíacos/metabolismo , Fatores de Regulação Miogênica/fisiologia , Proteínas com Domínio T/fisiologia
19.
Dev Biol ; 302(1): 208-17, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17054936

RESUMO

Although the signaling molecules regulating the early stages of valvular development have been well described, little is known on the late steps leading to mature fibrous leaflets. We hypothesized that atrioventricular (AV) valve development continues after birth to adjust to the postnatal maturation of the heart. By doing a systematic analysis of the AV valves of mice from embryonic day (E) 15.5 to 8 weeks old, we identified key developmental steps that map the maturation process of embryonic cushion-like leaflets into adult stress-resistant valves. Condensation of the mesenchymal cells occurred between E15.5 and E18.5 and was accompanied by increased cellular proliferation and adhesion. Cellular proliferation also contributed transiently to the concomitant elongation of the leaflets. Patterning of the extracellular matrix (ECM) proteins along the AV axis was achieved 1 week after birth, with the differentiation of two reciprocal structural regions, glycosaminoglycans and versican at the atrial side, and densely packed collagen fibers at the ventricular side. Formation and remodeling of the nodular thickenings at the closure points of the leaflets occurred between N4.5 and N11.5. In conclusion, AV valve development during late embryonic and postnatal stages includes condensation, elongation, formation of nodular thickenings, and remodeling of tension-resistant ECM proteins.


Assuntos
Matriz Extracelular/metabolismo , Valvas Cardíacas/crescimento & desenvolvimento , Valvas Cardíacas/metabolismo , Animais , Animais Recém-Nascidos , Contagem de Células , Proliferação de Células , Embrião de Mamíferos/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Coração/embriologia , Coração/crescimento & desenvolvimento , Septos Cardíacos/embriologia , Septos Cardíacos/crescimento & desenvolvimento , Valvas Cardíacas/embriologia , Mesoderma/citologia , Camundongos , Camundongos Endogâmicos
20.
Genesis ; 32(3): 218-30, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11892011

RESUMO

For nearly 40 years functional studies of the mouse quaking gene (qkI) have focused on its role in the postnatal central nervous system during myelination. However, the homozygous lethality of a number of ENU-induced alleles reveals that quaking has a critical role in embryonic development prior to the start of myelination. In this article, we show that quaking has a previously unsuspected and essential role in blood vessel development. Interestingly, we found that quaking, a nonsecreted protein, is expressed in the yolk sac endoderm, adjacent to the mesodermal site of developing blood islands, where the differentiation of blood and endothelial cells first occurs. Antibodies against PE-CAM-1, TIE-2 and SM-alpha-actin reveal that embryos homozygous for the qk(k2) allele have defective yolk sac vascular remodeling and abnormal vessels in the embryo proper at midgestation, coinciding with the timing of embryonic death. However, these mutants exhibit normal expression of Nkx2.5 and alpha-sarcomeric actin, indicating that cardiac muscle differentiation was normal. Further, they had normal embryonic heart rates in culture, suggesting that cardiac function was not compromised at this stage of embryonic development. Together, these results suggest that quaking plays an essential role in vascular development and that the blood vessel defects are the cause of embryonic death.


Assuntos
Vasos Sanguíneos/embriologia , Proteínas de Ligação a RNA/genética , Saco Vitelino/embriologia , Animais , Vasos Sanguíneos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Camundongos , Morfogênese , Proteínas de Ligação a RNA/fisiologia , Saco Vitelino/irrigação sanguínea , Saco Vitelino/fisiologia , Saco Vitelino/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA