Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Cell Sci ; 126(Pt 21): 4843-9, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24006259

RESUMO

Because nutrient-sensing nuclear and cytosolic acetylation mediates cellular autophagy, we investigated whether mitochondrial acetylation modulates mitochondrial autophagy (mitophagy). Knockdown of GCN5L1, a component of the mitochondrial acetyltransferase machinery, diminished mitochondrial protein acetylation and augmented mitochondrial enrichment of autophagy mediators. This program was disrupted by SIRT3 knockdown. Chronic GCN5L1 depletion increased mitochondrial turnover and reduced mitochondrial protein content and/or mass. In parallel, mitochondria showed blunted respiration and enhanced 'stress-resilience'. Genetic disruption of autophagy mediators Atg5 and p62 (also known as SQSTM1), as well as GCN5L1 reconstitution, abolished deacetylation-induced mitochondrial autophagy. Interestingly, this program is independent of the mitophagy E3-ligase Parkin (also known as PARK2). Taken together, these data suggest that deacetylation of mitochondrial proteins initiates mitochondrial autophagy in a canonical autophagy-mediator-dependent program and shows that modulation of this regulatory program has ameliorative mitochondrial homeostatic effects.


Assuntos
Autofagia , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Acetilação , Animais , Células HEK293 , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/enzimologia , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
2.
J Biol Chem ; 286(46): 40184-92, 2011 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-21930693

RESUMO

The mitochondrial permeability transition pore (mPTP) opening plays a critical role in mediating cell death during ischemia/reperfusion (I/R) injury. Our previous studies have shown that cysteine 203 of cyclophilin D (CypD), a critical mPTP mediator, undergoes protein S-nitrosylation (SNO). To investigate the role of cysteine 203 in mPTP activation, we mutated cysteine 203 of CypD to a serine residue (C203S) and determined its effect on mPTP opening. Treatment of WT mouse embryonic fibroblasts (MEFs) with H(2)O(2) resulted in an 50% loss of the mitochondrial calcein fluorescence, suggesting substantial activation of the mPTP. Consistent with the reported role of CypD in mPTP activation, CypD null (CypD(-/-)) MEFs exhibited significantly less mPTP opening. Addition of a nitric oxide donor, GSNO, to WT but not CypD(-/-) MEFs prior to H(2)O(2) attenuated mPTP opening. To test whether Cys-203 is required for this protection, we infected CypD(-/-) MEFs with a C203S-CypD vector. Surprisingly, C203S-CypD reconstituted MEFs were resistant to mPTP opening in the presence or absence of GSNO, suggesting a crucial role for Cys-203 in mPTP activation. To determine whether mutation of C203S-CypD would alter mPTP in vivo, we injected a recombinant adenovirus encoding C203S-CypD or WT CypD into CypD(-/-) mice via tail vein. Mitochondria isolated from livers of CypD(-/-) mice or mice expressing C203S-CypD were resistant to Ca(2+)-induced swelling as compared with WT CypD-reconstituted mice. Our results indicate that the Cys-203 residue of CypD is necessary for redox stress-induced activation of mPTP.


Assuntos
Ciclofilinas/metabolismo , Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Membranas Mitocondriais/metabolismo , Substituição de Aminoácidos , Animais , Cálcio/metabolismo , Células Cultivadas , Peptidil-Prolil Isomerase F , Ciclofilinas/genética , Cisteína , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Peróxido de Hidrogênio/farmacologia , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Proteínas de Transporte da Membrana Mitocondrial/genética , Poro de Transição de Permeabilidade Mitocondrial , Mutação de Sentido Incorreto , Doadores de Óxido Nítrico , Oxidantes/farmacologia , Oxirredução/efeitos dos fármacos
3.
Dev Dyn ; 240(9): 2127-41, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21780244

RESUMO

Although the function of transforming growth factor beta2 (TGFß2) in epithelial mesenchymal transition (EMT) is well studied, its role in valve remodeling remains to be fully explored. Here, we used histological, morphometric, immunohistochemical and molecular approaches and showed that significant dysregulation of major extracellular matrix (ECM) components contributed to valve remodeling defects in Tgfb2(-/-) embryos. The data indicated that cushion mesenchymal cell differentiation was impaired in Tgfb2(-/-) embryos. Hyaluronan and cartilage link protein-1 (CRTL1) were increased in hyperplastic valves of Tgfb2(-/-) embryos, indicating increased expansion and diversification of cushion mesenchyme into the cartilage cell lineage during heart development. Finally, Western blot and immunohistochemistry analyses indicate that the activation of SMAD2/3 was decreased in Tgfb2(-/-) embryos during valve remodeling. Collectively, the data indicate that TGFß2 promotes valve remodeling and differentiation by inducing matrix organization and suppressing cushion mesenchyme differentiation into cartilage cell lineage during heart development.


Assuntos
Valvas Cardíacas/metabolismo , Coração/embriologia , Fator de Crescimento Transformador beta2/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Matriz Extracelular/metabolismo , Valvas Cardíacas/embriologia , Imuno-Histoquímica , Mesoderma/citologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta2/genética
4.
J Mol Cell Cardiol ; 49(4): 693-8, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20600099

RESUMO

Prolonged cardiac overexpression of the mitochondrial biogenesis regulatory transcriptional coactivator PGC-1alpha disrupts cardiac contractile function and its genetic ablation limits cardiac capacity to enhance workload. In contrast, transient induction of PGC-1alpha alleviates neuronal cell oxidative stress and enhances skeletal myotube anti-oxidant defenses. We explored whether transient upregulation of PGC-1alpha in the heart protects against ischemia-reperfusion injury. The transient induction of PGC-1alpha in the cardiac-restricted inducible PGC-1alpha transgenic mouse, increased PGC-1alpha protein levels 5-fold. Following 25 min of ischemia and 2h of reperfusion on a Langendorff perfusion apparatus, contractile recovery and the rate pressure product was significantly blunted in mice overexpressing PGC-1alpha vs. controls. Affymetrix gene array analysis showed a 3-fold PGC-1alpha-mediated upregulation of adenine nucleotide translocase 1 (ANT1). As ANT1 upregulation induces cardiomyocyte cell death we investigated whether the induction of ANT1 by PGC-1alpha contributes to this enhanced ischemia-stress susceptibility. Infection with adenovirus harboring PGC-1alpha into cardiac-derived H9c2 cells significantly upregulates ANT1 without changing basal cell viability. In response to anoxia-reoxygenation injury cell death is significantly increased following PGC-1alpha overexpression. This detrimental effect is abolished following siRNA knockdown of ANT1. Similarly, the attenuation of ANT-1 in the presence of PGC-1alpha overexpression preserves the mitochondrial membrane potential in response to hydrogen-peroxide stress. Interestingly, the isolated knockdown of ANT1 also protects H9c2 cells from anoxia-reoxygenation injury. Taken together these data suggest that transient induction of PGC-1alpha in the murine heart decreases ischemia-reperfusion contractile recovery and diminishes anoxia-reoxygenation tolerance in H9c2 cells. These adverse phenotypes appear to be mediated, in part, by PGC-1alpha induced upregulation of ANT1.


Assuntos
Translocador 1 do Nucleotídeo Adenina/metabolismo , Traumatismo por Reperfusão/metabolismo , Transativadores/metabolismo , Translocador 1 do Nucleotídeo Adenina/genética , Animais , Sobrevivência Celular/fisiologia , Células Cultivadas , Citometria de Fluxo , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Camundongos Transgênicos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ratos , Traumatismo por Reperfusão/genética , Transativadores/genética , Fatores de Transcrição
5.
Circ Res ; 103(12): 1430-40, 2008 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-19008476

RESUMO

Congenital heart defects occur at a rate of 5% and are the most prevalent birth defects. A better understanding of the complex signaling networks regulating heart development is necessary to improve repair strategies for congenital heart defects. The mitogen-activated protein 3 kinase (MEKK3) is important to early embryogenesis, but developmental processes affected by MEKK3 during heart morphogenesis have not been fully examined. We identify MEKK3 as a critical signaling molecule during endocardial cushion development. We report the detection of MEKK3 transcripts to embryonic hearts before, during, and after cardiac cushion cells have executed epithelial-to-mesenchymal transition (EMT). MEKK3 is observed to endocardial cells of the cardiac cushions with a diminishing gradient of expression into the cushions. These observations suggest that MEKK3 may function during production of cushion mesenchyme as required for valvular development and septation of the heart. We used a kinase inactive form of MEKK3 (MEKK3(KI)) in an in vitro assay that recapitulates in vivo EMT and show that MEKK3(KI) attenuates mesenchyme formation. Conversely, constitutively active MEKK3 (ca-MEKK3) triggers mesenchyme production in ventricular endocardium, a tissue that does not normally undergo EMT. MEKK3-driven mesenchyme production is further substantiated by increased expression of EMT-relevant genes, including TGFbeta(2), Has2, and periostin. Furthermore, we show that MEKK3 stimulates EMT via a TGFbeta(2)-dependent mechanism. Thus, the activity of MEKK3 is sufficient for developmental EMT in the heart. This knowledge provides a basis to understand how MEKK3 integrates signaling cascades activating endocardial cushion EMT.


Assuntos
Coxins Endocárdicos/embriologia , Coxins Endocárdicos/enzimologia , Células Epiteliais/citologia , Células Epiteliais/enzimologia , MAP Quinase Quinase Quinase 3/fisiologia , Mesoderma/embriologia , Morfogênese/fisiologia , Fator de Crescimento Transformador beta2/fisiologia , Animais , Diferenciação Celular/fisiologia , Coxins Endocárdicos/citologia , Coxins Endocárdicos/metabolismo , Células Epiteliais/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , MAP Quinase Quinase Quinase 3/deficiência , MAP Quinase Quinase Quinase 3/genética , MAP Quinase Quinase Quinase 3/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos
6.
Cancer Res ; 65(15): 6755-63, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16061657

RESUMO

Metastatic invasion is the primary cause of breast cancer mortality, and adhesion receptors, such as CD44, are believed to be critical in this process. Historically, primary breast tumor epithelium has been investigated in isolation from other tissue components, leading to the common interpretation that CD44 and its primary ligand, hyaluronan, promote invasion. Here, we provide in vivo evidence showing CD44 antagonism to breast cancer metastasis. In a mouse model of spontaneously metastasizing breast cancer (MMTV-PyV mT), we found that loss of CD44 promotes metastasis to the lung. Localization studies, in combination with a novel hyaluronan synthase-GFP transgenic mouse, show a restricted pattern of expression for CD44 and hyaluronan. Whereas CD44 is expressed in tumor epithelium, hyaluronan synthase expression is restricted to stromal-associated cells. This distinct CD44 and hyaluronan pattern of distribution suggests a role for epithelial-stromal interaction in CD44 function. To define the relevance of this spatial regulation, we developed an in vitro invasion assay to emulate invasion into the extracellular matrix. Invasion of CD44-positive tumor cells was inhibited in hyaluronan-containing matrices, whereas blocking CD44-hyaluronan association increased invasion. Collectively, these data show that during breast cancer progression, hyaluronan-CD44 dynamics occurring through epithelial-stromal interactions are protective against metastasis.


Assuntos
Neoplasias da Mama/patologia , Receptores de Hialuronatos/fisiologia , Ácido Hialurônico/fisiologia , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Progressão da Doença , Feminino , Proteínas de Fluorescência Verde/biossíntese , Humanos , Receptores de Hialuronatos/biossíntese , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/metabolismo , Neoplasias Pulmonares/prevenção & controle , Masculino , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Invasividade Neoplásica
7.
Sci Rep ; 7(1): 2093, 2017 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-28522833

RESUMO

The regulatory control of cardiac endoplasmic reticulum (ER) stress is incompletely characterized. As ER stress signaling upregulates the E3-ubiquitin ligase Parkin, we investigated the role of Parkin in cardiac ER stress. Parkin knockout mice exposed to aortic constriction-induced cardiac pressure-overload or in response to systemic tunicamycin (TM) developed adverse ventricular remodeling with excessive levels of the ER regulatory C/EBP homologous protein CHOP. CHOP was identified as a Parkin substrate and its turnover was Parkin-dose and proteasome-dependent. Parkin depletion in cardiac HL-1 cells increased CHOP levels and enhanced susceptibility to TM-induced cell death. Parkin reconstitution rescued this phenotype and the contribution of excess CHOP to this ER stress injury was confirmed by reduction in TM-induced cell death when CHOP was depleted in Parkin knockdown cardiomyocytes. Isogenic Parkin mutant iPSC-derived cardiomyocytes showed exaggerated ER stress induced CHOP and apoptotic signatures and myocardium from subjects with dilated cardiomyopathy showed excessive Parkin and CHOP induction. This study identifies that Parkin functions to blunt excessive CHOP to prevent maladaptive ER stress-induced cell death and adverse cardiac ventricular remodeling. Additionally, Parkin is identified as a novel post-translational regulatory moderator of CHOP stability and uncovers an additional stress-modifying function of this E3-ubiquitin ligase.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Estresse do Retículo Endoplasmático , Miócitos Cardíacos/metabolismo , Fator de Transcrição CHOP/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Apoptose , Cardiomiopatia Dilatada/patologia , Linhagem Celular , Linhagem Celular Tumoral , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/citologia , Ubiquitina-Proteína Ligases/genética , Remodelação Ventricular
8.
Gene Expr Patterns ; 6(5): 462-70, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16458617

RESUMO

Heart septation and valve malformations constitute the most common birth defects. These cardiac structures arise from the endocardial cushions through dynamic interactions between cells and the extracellular matrix (cardiac jelly). Targeted deletion of the hyaluronan synthase-2 (Has2) gene in mice results in an absence of cardiac jelly and endocardial cushions, a loss of vascular integrity, and embryonic death at E9.5. Despite the requirements for Has2 and its synthetic product hyaluronan (HA) in the developing cardiovascular system, little is known about the normal expression pattern of Has2 or the factors regulating Has2 gene transcription during development. Bmp signaling is an important regulator of cardiac myogenesis, and is also important for endocardial cushion formation. The current study defines the embryonic expression pattern of Has2 and explores the regulation of Has2 gene expression by Bmp signaling. In situ hybridization studies demonstrate dynamic Has2 expression patterns during myocardial cell development and cardiac tube formation, formation of the cardiac endocardial cushions, and cushion invasion by valve primordial cells. Despite overlapping regional expression of Bmp2 in the late gastrula anterior lateral endoderm and Has2 in the adjacent cardiogenic mesoderm, application of noggin-expressing CHO cells beneath the endoderm failed to perturb normal Has2 expression. Thus, in contrast to many genes expressed in the heart forming region, regulation of Has2 in the cardiogenic mesoderm is independent of Bmp signaling.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Glucuronosiltransferase/genética , Coração/embriologia , Transdução de Sinais , Animais , Células CHO , Embrião de Galinha , Cricetinae , Hialuronan Sintases , Camundongos , Morfogênese
9.
J Clin Invest ; 121(9): 3701-12, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21865652

RESUMO

It has long been hypothesized that abnormalities in lipid biology contribute to degenerative brain diseases. Consistent with this, emerging epidemiologic evidence links lipid alterations with Parkinson disease (PD), and disruption of lipid metabolism has been found to predispose to α-synuclein toxicity. We therefore investigated whether Parkin, an E3 ubiquitin ligase found to be defective in patients with early onset PD, regulates systemic lipid metabolism. We perturbed lipid levels by exposing Parkin+/+ and Parkin-/- mice to a high-fat and -cholesterol diet (HFD). Parkin-/- mice resisted weight gain, steatohepatitis, and insulin resistance. In wild-type mice, the HFD markedly increased hepatic Parkin levels in parallel with lipid transport proteins, including CD36, Sr-B1, and FABP. These lipid transport proteins were not induced in Parkin-/- mice. The role of Parkin in fat uptake was confirmed by increased oleate accumulation in hepatocytes overexpressing Parkin and decreased uptake in Parkin-/- mouse embryonic fibroblasts and patient cells harboring complex heterozygous mutations in the Parkin-encoding gene PARK2. Parkin conferred this effect, in part, via ubiquitin-mediated stabilization of the lipid transporter CD36. Reconstitution of Parkin restored hepatic fat uptake and CD36 levels in Parkin-/- mice, and Parkin augmented fat accumulation during adipocyte differentiation. These results demonstrate that Parkin is regulated in a lipid-dependent manner and modulates systemic fat uptake via ubiquitin ligase-dependent effects. Whether this metabolic regulation contributes to premature Parkinsonism warrants investigation.


Assuntos
Gorduras na Dieta/metabolismo , Metabolismo dos Lipídeos , Ubiquitina-Proteína Ligases/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Temperatura Corporal , Antígenos CD36/genética , Antígenos CD36/metabolismo , Linhagem Celular , Ingestão de Alimentos , Metabolismo Energético , Glucose/metabolismo , Humanos , Insulina/sangue , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Ubiquitina-Proteína Ligases/genética , Aumento de Peso , alfa-Sinucleína/metabolismo
10.
J Angiogenes Res ; 1: 2, 2009 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-19946410

RESUMO

Endothelial to mesenchyme transition (EndMT) can be observed during the formation of endocardial cushions from the endocardium, the endothelial lining of the atrioventricular canal (AVC), of the developing heart at embryonic day 9.5 (E9.5). Many regulators of the process have been identified; however, the mechanisms driving the initial commitment decision of endothelial cells to EndMT have been difficult to separate from processes required for mesenchymal proliferation and migration. We have several lines of evidence that suggest a central role for Akt signaling in committing endothelial cells to enter EndMT. Akt1 mRNA was restricted to the endocardium of endocardial cushions while they were forming. The PI3K/Akt signaling pathway is necessary for mesenchyme outgrowth, as sprouting was inhibited in AVC explant cultures treated with the PI3K inhibitor LY294002. Furthermore, endothelial marker, VE-cadherin, was downregulated and mesenchyme markers, N-cadherin and Snail, were induced in response to expression of a constitutively active form of Akt1 (myrAkt1) in endothelial cells. Finally, we isolated the function of Akt1 signaling in the commitment to the transition using a transgenic model where myrAkt1 was pulsed only in endocardial cells and turned off after EndMT initiation. In this way, we determined that increased Akt signaling in the endocardium drives EndMT and discounted its other functions in cushion mesenchymal cells.

11.
Dev Dyn ; 237(11): 3102-14, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18855897

RESUMO

The cytoplasmic serine/threonine kinases transduce extracellular signals into regulatory events that impact cellular responses. The induction of one kinase triggers the activation of several downstream kinases, leading to the regulation of transcription factors to affect gene function. This arrangement allows for the kinase cascade to be amplified, and integrated according to the cellular context. An upstream mitogen or growth factor signal initiates a module of three kinases: a mitogen-activated protein (MAP) kinase kinase kinase (MAPKKK; e.g., Raf) that phosphorylates and activates a MAP kinase kinase (MAPKK; e.g., MEK) and finally activation of MAP kinase (MAPK; e.g., ERK). Thus, this MAP3K-MAP2K-MAPK module represents critical effectors that regulate extracellular stimuli into cellular responses, such as differentiation, proliferation, and apoptosis all of which function during development. There are 21 characterized MAP3Ks that activate known MAP2Ks, and they function in many aspects of developmental biology. This review summarizes known transduction routes linked to each MAP3K and highlights mouse models that provide clues to their physiological functions. This perspective reveals that some of these MAP3K effectors may have redundant functions, and also serve as unique nexus depending on the context of the signaling pathway.


Assuntos
MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Modelos Biológicos , Animais , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Mitógenos/metabolismo
12.
Dev Dyn ; 235(10): 2761-70, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16894626

RESUMO

Congenital heart malformations occur at a rate of one per one hundred births and are considered the most frequent birth defects. This high incidence of cardiac defects underscores the complex developmental processes required to form the first functioning organ in mammals. The molecular cues which govern heart development are poorly defined and require an improved understanding in order to advance repair strategies for heart defects. The cytoplasmic MAP kinase kinase kinase, MEKK4, is a critical effector in cellular stress responses; however, the function of MEKK4 during embryonic development and cardiogenesis is not well understood. We have identified MEKK4 as a critical signaling molecule during cardiovascular development. We report the detection of MEKK4 transcripts to early myocardium, endocardium and to cardiac cushion cells that have executed epithelial to mesenchymal transformation (EMT). These observations suggest that MEKK4 may function during production of the cushion mesenchyme as required to create valves and the septated heart. We used a kinase inactive form of MEKK4(MEKK4(KI)) in an in vitro assay that recapitulates in vivo EMT, and show that MEKK4(KI) attenuates mesenchyme production. However, addition of a constitutively active MEKK4 into ventricular explants, a system that does not normally undergo EMT, is not able to cause mesenchymal cell outgrowth. Thus, the kinase activity of MEKK4 is essential, but not sufficient, to support developmental EMT. This knowledge provides a basis to understand how MEKK4 may integrate signaling cascades controlling heart development.


Assuntos
Células Epiteliais/metabolismo , Ventrículos do Coração/metabolismo , MAP Quinase Quinase Quinase 4/genética , Mesoderma/metabolismo , Animais , Western Blotting , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Ventrículos do Coração/embriologia , Ventrículos do Coração/enzimologia , Imuno-Histoquímica , Imunoprecipitação , Hibridização In Situ/métodos , Técnicas In Vitro , MAP Quinase Quinase Quinase 4/metabolismo , Mesoderma/citologia , Mesoderma/enzimologia , Camundongos , Organogênese/genética , Organogênese/fisiologia , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA