Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cardiovasc Res ; 118(16): 3225-3238, 2022 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-35104324

RESUMO

AIMS: Pulmonary arterial hypertension (PAH) is a devastating disease with limited therapeutic options. Vascular remodelling of pulmonary arteries, characterized by increased proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs), is a hallmark of PAH. Here, we aimed to systematically characterize coagulation-independent effects of key coagulation proteases thrombin and Factor Xa (FXa) and their designated receptors, protease-activated receptor (PAR)-1 and -2, on PASMCs in vitro and experimental PAH in vivo. METHODS AND RESULTS: In human and murine PASMCs, both thrombin and FXa were identified as potent mitogens, and chemoattractants. FXa mediated its responses via PAR-1 and PAR-2, whereas thrombin signalled through PAR-1. Extracellular-signal regulated kinases 1/2, protein kinase B (AKT), and sphingosine kinase 1 were identified as downstream mediators of PAR-1 and PAR-2. Inhibition of FXa or thrombin blunted cellular responses in vitro, but unexpectedly failed to protect against hypoxia-induced PAH in vivo. However, pharmacological inhibition as well as genetic deficiency of both PAR-1 and PAR-2 significantly reduced vascular muscularization of small pulmonary arteries, diminished right ventricular systolic pressure, and right ventricular hypertrophy upon chronic hypoxia compared to wild-type controls. CONCLUSION: Our findings indicate a coagulation-independent pathogenic potential of thrombin and FXa for pulmonary vascular remodelling via acting through PAR-1 and PAR-2, respectively. While inhibition of single coagulation proteases was ineffective in preventing experimental PAH, our results propose a crucial role for PAR-1 and PAR-2 in its pathobiology, thus identifying PARs but not their dedicated activators FXa and thrombin as suitable targets for the treatment of PAH.


Assuntos
Hipertensão Pulmonar , Trombina , Camundongos , Humanos , Animais , Trombina/metabolismo , Fator Xa/metabolismo , Fator Xa/farmacologia , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/prevenção & controle , Remodelação Vascular , Receptor PAR-1/genética , Receptor PAR-2/genética , Receptor PAR-2/metabolismo , Hipóxia
2.
J Clin Invest ; 131(19)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34596056

RESUMO

Enhanced signaling via RTKs in pulmonary hypertension (PH) impedes current treatment options because it perpetuates proliferation and apoptosis resistance of pulmonary arterial smooth muscle cells (PASMCs). Here, we demonstrated hyperphosphorylation of multiple RTKs in diseased human vessels and increased activation of their common downstream effector phosphatidylinositol 3'-kinase (PI3K), which thus emerged as an attractive therapeutic target. Systematic characterization of class IA catalytic PI3K isoforms identified p110α as the key regulator of pathogenic signaling pathways and PASMC responses (proliferation, migration, survival) downstream of multiple RTKs. Smooth muscle cell-specific genetic ablation or pharmacological inhibition of p110α prevented onset and progression of pulmonary hypertension (PH) as well as right heart hypertrophy in vivo and even reversed established vascular remodeling and PH in various animal models. These effects were attributable to both inhibition of vascular proliferation and induction of apoptosis. Since this pathway is abundantly activated in human disease, p110α represents a central target in PH.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Hipertensão Pulmonar/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase/uso terapêutico , Adulto , Animais , Células Cultivadas , Humanos , Hipertensão Pulmonar/etiologia , Lactente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
3.
Front Immunol ; 12: 701721, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34691017

RESUMO

The six-transmembrane protein of prostate 2 (Stamp2) acts as an anti-inflammatory protein in macrophages by protecting from overt inflammatory signaling and Stamp2 deficiency accelerates atherosclerosis in mice. Herein, we describe an unexpected role of Stamp2 in polymorphonuclear neutrophils (PMN) and characterize Stamp2's protective effects in myocardial ischemic injury. In a murine model of ischemia and reperfusion (I/R), echocardiography and histological analyses revealed a pronounced impairment of cardiac function in hearts of Stamp2-deficient- (Stamp2-/- ) mice as compared to wild-type (WT) animals. This difference was driven by aggravated cardiac fibrosis, as augmented fibroblast-to-myofibroblast transdifferentiation was observed which was mediated by activation of the redox-sensitive p38 mitogen-activated protein kinase (p38 MAPK). Furthermore, we observed increased production of reactive oxygen species (ROS) in Stamp2-/- hearts after I/R, which is the likely cause for p38 MAPK activation. Although myocardial macrophage numbers were not affected by Stamp2 deficiency after I/R, augmented myocardial infiltration by polymorphonuclear neutrophils (PMN) was observed, which coincided with enhanced myeloperoxidase (MPO) plasma levels. Primary PMN isolated from Stamp2-/- animals exhibited a proinflammatory phenotype characterized by enhanced nuclear factor (NF)-κB activity and MPO secretion. To prove the critical role of PMN for the observed phenotype after I/R, antibody-mediated PMN depletion was performed in Stamp2-/- mice which reduced deterioration of LV function and adverse structural remodeling to WT levels. These data indicate a novel role of Stamp2 as an anti-inflammatory regulator of PMN and fibroblast-to-myofibroblast transdifferentiation in myocardial I/R injury.


Assuntos
Coração/fisiologia , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Miocárdio/metabolismo , Animais , Cardiomiopatias/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , NF-kappa B/metabolismo , Ativação de Neutrófilo/fisiologia , Neutrófilos/metabolismo , Peroxidase/metabolismo , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
4.
Basic Res Cardiol ; 115(6): 68, 2020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-33188479

RESUMO

Six-transmembrane protein of prostate (Stamp2) protects from diabetes and atherosclerosis in mice via anti-inflammatory mechanisms. As chronic inflammation is a hallmark of pulmonary arterial hypertension (PAH), we investigated the role of Stamp2. Stamp2 expression was substantially reduced in the lung of humans with idiopathic PAH, as well as in experimental PAH. In Stamp2-deficient mice, hypoxia modestly aggravated pulmonary vascular remodeling and right ventricular pressure compared to WT. As endothelial cell (EC) and pulmonary arterial smooth muscle cell (PASMC) phenotypes drive remodeling in PAH, we explored the role of Stamp2. Knock-down of Stamp2 in human EC neither affected apoptosis, viability, nor release of IL-6. Moreover, Stamp2 deficiency in primary PASMC did not alter mitogenic or migratory properties. As Stamp2 deficiency augmented expression of inflammatory cytokines and numbers of CD68-positive cells in the lung, actions of Stamp2 in macrophages may drive vascular remodeling. Thus, PASMC responses were assessed following treatment with conditioned media of primary Stamp2-/- or WT macrophages. Stamp2-/- supernatants induced PASMC proliferation and migration stronger compared to WT. A cytokine array revealed CXCL12, MCP-1 and IL-6 as most relevant candidates. Experiments with neutralizing antibodies confirmed the role of these cytokines in driving Stamp2's responses. In conclusion, Stamp2 deficiency aggravates pulmonary vascular remodeling via cross-talk between macrophages and PASMC. Despite a substantial pro-inflammatory response, the hemodynamic effect of Stamp2 deficiency is modest suggesting that additional mechanisms apart from inflammation are necessary to induce severe PAH.


Assuntos
Hipertensão Pulmonar/metabolismo , Proteínas de Membrana/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Remodelação Vascular , Adolescente , Adulto , Animais , Comunicação Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Pré-Escolar , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Humanos , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Hipóxia/complicações , Lactente , Macrófagos/metabolismo , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos Knockout , Pessoa de Meia-Idade , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/patologia , Oxirredutases/genética , Oxirredutases/metabolismo , Pneumonia/etiologia , Pneumonia/metabolismo , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Artéria Pulmonar/fisiopatologia , Ratos Sprague-Dawley , Transdução de Sinais , Disfunção Ventricular Direita/etiologia , Disfunção Ventricular Direita/metabolismo , Disfunção Ventricular Direita/fisiopatologia , Função Ventricular Direita
5.
Arterioscler Thromb Vasc Biol ; 35(6): 1434-44, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25908763

RESUMO

OBJECTIVE: Neointima formation after vascular injury remains a significant problem in clinical cardiology, and current preventive strategies are suboptimal. Phosphatidylinositol 3'-kinase is a central downstream mediator of growth factor signaling, but the role of phosphatidylinositol 3'-kinase isoforms in vascular remodeling remains elusive. We sought to systematically characterize the precise role of catalytic class IA phosphatidylinositol 3'-kinase isoforms (p110α, p110ß, p110δ), which signal downstream of receptor tyrosine kinases, for vascular remodeling in vivo. APPROACH AND RESULTS: Western blot analyses revealed that all 3 isoforms are abundantly expressed in smooth muscle cells. To analyze their significance for receptor tyrosine kinases-dependent cellular responses, we used targeted gene knockdown and isoform-specific small molecule inhibitors of p110α (PIK-75), p110ß (TGX-221), and p110δ (IC-87114), respectively. We identified p110α to be crucial for receptor tyrosine kinases signaling, thus affecting proliferation, migration, and survival of rat, murine, and human smooth muscle cells, whereas p110ß and p110δ activities were dispensable. Surprisingly, p110δ exerted noncatalytic functions in smooth muscle cell proliferation, but had no effect on migration. Based on these results, we generated a mouse model of smooth muscle cell-specific p110α deficiency (sm-p110α(-/-)). Targeted deletion of p110α in sm-p110α(-/-) mice blunted growth factor-induced cellular responses and abolished neointima formation after balloon injury of the carotid artery in mice. In contrast, p110δ deficiency did not affect vascular remodeling in vivo. CONCLUSIONS: Receptor tyrosine kinases-induced phosphatidylinositol 3'-kinase signaling via the p110α isoform plays a central role for vascular remodeling in vivo. Thus, p110α represents a selective target for the prevention of neointima formation after vascular injury, whereas p110ß and p110δ expression and activity do not play a significant role.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Remodelação Vascular/fisiologia , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Classe Ia de Fosfatidilinositol 3-Quinase/farmacologia , Humanos , Camundongos , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/enzimologia , Neointima/prevenção & controle , Isoformas de Proteínas , Ratos , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais
6.
Arterioscler Thromb Vasc Biol ; 35(5): 1236-45, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25745058

RESUMO

OBJECTIVE: Despite modern therapies, pulmonary arterial hypertension (PAH) harbors a high mortality. Vascular remodeling is a hallmark of the disease. Recent clinical studies revealed that antiremodeling approaches with tyrosine-kinase inhibitors such as imatinib are effective, but its applicability is limited by significant side effects. Although imatinib has multiple targets, expression analyses support a role for platelet-derived growth factor (PDGF) in the pathobiology of the disease. However, its precise role and downstream signaling events have not been established. APPROACH AND RESULTS: Patients with PAH exhibit enhanced expression and phosphorylation of ß PDGF receptor (ßPDGFR) in remodeled pulmonary arterioles, particularly at the binding sites for phophatidyl-inositol-3-kinase and PLCγ at tyrosine residues 751 and 1021, respectively. These signaling molecules were identified as critical downstream mediators of ßPDGFR-mediated proliferation and migration of pulmonary arterial smooth muscle cells. We, therefore, investigated mice expressing a mutated ßPDGFR that is unable to recruit phophatidyl-inositol-3-kinase and PLCγ (ßPDGFR(F3/F3)). PDGF-dependent Erk1/2 and Akt phosphorylation, cyclin D1 induction, and proliferation, migration, and protection against apoptosis were abolished in ßPDGFR(F3/F3) pulmonary arterial smooth muscle cells. On exposure to chronic hypoxia, vascular remodeling of pulmonary arteries was blunted in ßPDGFR(F3/F3) mice compared with wild-type littermates. These alterations led to protection from hypoxia-induced PAH and right ventricular hypertrophy. CONCLUSIONS: By means of a genetic approach, our data provide definite evidence that the activated ßPDGFR is a key contributor to pulmonary vascular remodeling and PAH. Selective disruption of PDGF-dependent phophatidyl-inositol-3-kinase and PLCγ activity is sufficient to abolish these pathogenic responses in vivo, identifying these signaling events as valuable targets for antiremodeling strategies in PAH.


Assuntos
Hipertensão Pulmonar/genética , Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais/genética , Remodelação Vascular/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Hipertensão Pulmonar/patologia , Camundongos , Mutação , Fator de Crescimento Derivado de Plaquetas/metabolismo , Sensibilidade e Especificidade , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Expert Opin Investig Drugs ; 21(1): 119-34, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22074410

RESUMO

INTRODUCTION: Despite recent advances, pulmonary arterial hypertension (PAH) remains a devastating disease which harbors a poor prognosis. Novel therapeutic approaches directly targeting pulmonary vascular remodeling are warranted. AREAS COVERED: This review delineates the current limitations in the management of PAH and focuses on a novel, anti-proliferative therapeutic concept. It will help readers understand the mechanisms of receptor tyrosine kinase signaling, with a special focus on platelet-derived growth factor (PDGF) receptors and their role in the pathobiology of PAH. Furthermore, it provides a comprehensive summary regarding the rationale, efficacy and safety of the tyrosine kinase inhibitor imatinib mesylate , which potently inhibits the PDGF receptor, as an additional treatment option in PAH. EXPERT OPINION: PDGF is a potent mitogen for pulmonary vascular smooth muscle cells and represents an important mediator of pulmonary vascular remodeling. Imatinib mesylate, a compound that inhibits the Bcr-Abl kinase and was developed for the treatment of chronic myeloid leukemia, also targets PDGF receptors. Both experimental and clinical data indicate that it reverses the vascular remodeling process even when it is fully established. Results from Phase II and III clinical trials suggest potent and prolonged efficacy in patients with severe PAH (i.e., pulmonary vascular resistance > 800 dynes*s*cm(-5)). Future studies should evaluate the long-term clinical efficacy and safety of imatinib, including patients with less impaired hemodynamics. Based on the current knowledge, this compound is likely to become an additional treatment option for patients with PAH and has the potential to at least partially correct the pathology of the disease.


Assuntos
Hipertensão Pulmonar/tratamento farmacológico , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Animais , Benzamidas , Ensaios Clínicos como Assunto , Sistemas de Liberação de Medicamentos , Humanos , Hipertensão Pulmonar/fisiopatologia , Mesilato de Imatinib , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Piperazinas/efeitos adversos , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/efeitos adversos , Pirimidinas/farmacologia , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Fatores de Tempo , Resultado do Tratamento
8.
PLoS One ; 6(11): e26628, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22125598

RESUMO

BACKGROUND: Neuroendocrine activation and local mediators such as transforming growth factor-ß1 (TGF-ß1) contribute to the pathobiology of cardiac hypertrophy and failure, but the underlying mechanisms are incompletely understood. We aimed to characterize the functional network involving TGF-ß1, the renin-angiotensin system, and the ß-adrenergic system in the heart. METHODS: Transgenic mice overexpressing TGF-ß1 (TGF-ß1-Tg) were treated with a ß-blocker, an AT1-receptor antagonist, or a TGF-ß-antagonist (sTGFßR-Fc), were morphologically characterized. Contractile function was assessed by dobutamine stress echocardiography in vivo and isolated myocytes in vitro. Functional alterations were related to regulators of cardiac energy metabolism. RESULTS: Compared to wild-type controls, TGF-ß1-Tg mice displayed an increased heart-to-body-weight ratio involving both fibrosis and myocyte hypertrophy. TGF-ß1 overexpression increased the hypertrophic responsiveness to ß-adrenergic stimulation. In contrast, the inotropic response to ß-adrenergic stimulation was diminished in TGF-ß1-Tg mice, albeit unchanged basal contractility. Treatment with sTGF-ßR-Fc completely prevented the cardiac phenotype in transgenic mice. Chronic ß-blocker treatment also prevented hypertrophy and ANF induction by isoprenaline, and restored the inotropic response to ß-adrenergic stimulation without affecting TGF-ß1 levels, whereas AT1-receptor blockade had no effect. The impaired contractile reserve in TGF-ß1-Tg mice was accompanied by an upregulation of mitochondrial uncoupling proteins (UCPs) which was reversed by ß-adrenoceptor blockade. UCP-inhibition restored the contractile response to ß-adrenoceptor stimulation in vitro and in vivo. Finally, cardiac TGF-ß1 and UCP expression were elevated in heart failure in humans, and UCP--but not TGF-ß1--was downregulated by ß-blocker treatment. CONCLUSIONS: Our data support the concept that TGF-ß1 acts downstream of angiotensin II in cardiomyocytes, and furthermore, highlight the critical role of the ß-adrenergic system in TGF-ß1-induced cardiac phenotype. Our data indicate for the first time, that TGF-ß1 directly influences mitochondrial energy metabolism by regulating UCP3 expression. ß-blockers may act beneficially by normalizing regulatory mechanisms of cellular hypertrophy and energy metabolism.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Cardiomegalia/metabolismo , Coração/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo , Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Benzimidazóis/farmacologia , Benzoatos/farmacologia , Cardiomegalia/diagnóstico por imagem , Cardiomegalia/genética , Células Cultivadas , Ecocardiografia sob Estresse , Regulação da Expressão Gênica/efeitos dos fármacos , Coração/fisiologia , Humanos , Canais Iônicos/genética , Isoproterenol/farmacologia , Metoprolol/farmacologia , Camundongos , Camundongos Transgênicos , Proteínas Mitocondriais/genética , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Telmisartan , Fator de Crescimento Transformador beta1/genética , Proteína Desacopladora 3
9.
J Am Coll Cardiol ; 57(25): 2527-38, 2011 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-21679854

RESUMO

OBJECTIVES: We tested the hypothesis whether selective blunting of platelet-derived growth factor (PDGF)-dependent vascular smooth muscle cell (VSMC) proliferation and migration is sufficient to prevent neointima formation after vascular injury. BACKGROUND: To prevent neointima formation and stent thrombosis after coronary interventions, it is essential to inhibit VSMC proliferation and migration without harming endothelial cell function. The role of PDGF-a potent mitogen and chemoattractant for VSMC that does not affect endothelial cells-for neointima formation remains controversial. METHODS: To decipher the signaling pathways that control PDGF beta receptor (ßPDGFR)-driven VSMC proliferation and migration, we characterized 2 panels of chimeric CSF1R/ßPDGFR mutants in which the binding sites for ßPDGFR-associated signaling molecules (Src, phosphatidylinositol 3-kinase [PI3K], GTPase activating protein of ras, SHP-2, phospholipase Cγ 1 [PLCγ]) were individually mutated. Based on in vitro results, the importance of PDGF-initiated signals for neointima formation was investigated in genetically modified mice. RESULTS: Our results indicate that the chemotactic response to PDGF requires the activation of Src, PI3K, and PLCγ, whereas PDGF-dependent cell cycle progression is exclusively mediated by PI3K and PLCγ. These 2 signaling molecules contribute to signal relay of the ßPDGFR by differentially regulating cyclin D1 and p27(kip1). Blunting of ßPDGFR-induced PI3K and PLCγ signaling by a combination mutant (F3) completely abolished the mitogenic and chemotactic response to PDGF. Disruption of PDGF-dependent PI3K and PLCγ signaling in mice expressing the F3 receptor led to a profound reduction of neointima formation after balloon injury. CONCLUSIONS: Signaling by the activated ßPDGFR, particularly through PI3K and PLCγ, is crucial for neointima formation after vascular injury. Disruption of these specific signaling pathways is sufficient to attenuate pathogenic processes such as vascular remodeling in vivo.


Assuntos
Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/patologia , Neointima/prevenção & controle , Fosfatidilinositol 3-Quinase/metabolismo , Fosfolipase C gama/metabolismo , Animais , Movimento Celular , Proliferação de Células , Camundongos , Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais
10.
Am J Respir Crit Care Med ; 183(8): 1092-102, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21177885

RESUMO

RATIONALE: Platelet-derived growth factor (PDGF) plays a pivotal role in the pathobiology of pulmonary hypertension (PH) because it promotes pulmonary vascular remodeling. PH is frequently associated with pulmonary hypoxia. OBJECTIVES: To investigate whether hypoxia alters PDGF ß receptor (ßPDGFR) signaling in the pulmonary vasculature. METHODS: The impact of chronic hypoxia on signal transduction by the ßPDGFR was measured in human pulmonary arterial smooth muscle cells (hPASMC) in vitro, and in mice with hypoxia-induced PH in vivo. MEASUREMENTS AND MAIN RESULTS: Chronic hypoxia significantly enhanced PDGF-BB-dependent proliferation and chemotaxis of hPASMC. Pharmacologic inhibition of PI3 kinase (PI3K) and PLCγ abrogated these events under both normoxia and hypoxia. Although hypoxia did not affect ßPDGFR expression, it increased the ligand-induced tyrosine phosphorylation of the receptor, particularly at binding sites for PI3K (Y751) and PLCγ (Y1021). The activated ßPDGFR is dephosphorylated by protein tyrosine phosphatases (PTPs). Interestingly, hypoxia decreased expression of numerous PTPs (T cell PTP, density-enhanced phosphatase-1, PTP1B, and SH2 domain-containing phosphatase-2), resulting in reduced PTP activity. Hypoxia-inducible factor (HIF)-1α is involved in this regulation of gene expression, because hypoxia-induced ßPDGFR hyperphosphorylation and PTP down-regulation were abolished by HIF-1α siRNA and by the HIF-1α inhibitor 2-methoxyestradiol. ßPDGFR hyperphosphorylation and PTP down-regulation were also present in vivo in mice with chronic hypoxia-induced PH. CONCLUSIONS: Hypoxia reduces expression and activity of ßPDGFR-antagonizing PTPs in a HIF-1α-dependent manner, thereby enhancing receptor activation and proliferation and chemotaxis of hPASMC. Because hyperphosphorylation of the ßPDGFR and down-regulation of PTPs occur in vivo, this mechanism likely has significant impact on the development and progression of PH and other hypoxia-associated diseases.


Assuntos
Hipóxia/fisiopatologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Proteínas Tirosina Fosfatases/fisiologia , Artéria Pulmonar/fisiopatologia , Animais , Proliferação de Células , Células Cultivadas , Quimiotaxia/fisiologia , Regulação para Baixo/fisiologia , Humanos , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/fisiopatologia , Pulmão/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/fisiopatologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Transdução de Sinais/fisiologia
11.
PLoS One ; 5(10): e13608, 2010 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-21049052

RESUMO

BACKGROUND: Profilin-1 is an ubiquitous actin binding protein. Under pathological conditions such as diabetes, profilin-1 levels are increased in the vascular endothelium. We recently demonstrated that profilin-1 overexpression triggers indicators of endothelial dysfunction downstream of LDL signaling, and that attenuated expression of profilin-1 confers protection from atherosclerosis in vivo. METHODOLOGY: Here we monitored profilin-1 expression in human atherosclerotic plaques by immunofluorescent staining. The effects of recombinant profilin-1 on atherogenic signaling pathways and cellular responses such as DNA synthesis (BrdU-incorporation) and chemotaxis (modified Boyden-chamber) were evaluated in cultured rat aortic and human coronary vascular smooth muscle cells (VSMCs). Furthermore, the correlation between profilin-1 serum levels and the degree of atherosclerosis was assessed in humans. PRINCIPAL FINDINGS: In coronary arteries from patients with coronary heart disease, we found markedly enhanced profilin expression in atherosclerotic plaques compared to the normal vessel wall. Stimulation of rat aortic and human coronary VSMCs with recombinant profilin-1 (10(-6) M) in vitro led to activation of intracellular signaling cascades such as phosphorylation of Erk1/2, p70(S6) kinase and PI3K/Akt within 10 minutes. Furthermore, profilin-1 concentration-dependently induced DNA-synthesis and migration of both rat and human VSMCs, respectively. Inhibition of PI3K (Wortmannin, LY294002) or Src-family kinases (SU6656, PP2), but not PLCγ (U73122), completely abolished profilin-induced cell cycle progression, whereas PI3K inhibition partially reduced the chemotactic response. Finally, we found that profilin-1 serum levels were significantly elevated in patients with severe atherosclerosis in humans (p<0.001 vs. no atherosclerosis or control group). CONCLUSIONS: Profilin-1 expression is significantly enhanced in human atherosclerotic plaques compared to the normal vessel wall, and the serum levels of profilin-1 correlate with the degree of atherosclerosis in humans. The atherogenic effects exerted by profilin-1 on VSMCs suggest an auto-/paracrine role within the plaque. These data indicate that profilin-1 might critically contribute to atherogenesis and may represent a novel therapeutic target.


Assuntos
Aterosclerose/patologia , Músculo Liso Vascular/metabolismo , Profilinas/metabolismo , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA , Humanos , Músculo Liso Vascular/patologia , Fosforilação , Reação em Cadeia da Polimerase , Profilinas/fisiologia , Ratos , Ratos Endogâmicos WKY , Transdução de Sinais
12.
J Mol Cell Cardiol ; 48(6): 1316-23, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20307544

RESUMO

Platelet-derived-growth-factor-BB (PDGF-BB) can protect various cell types from apoptotic cell death, and induce hypertrophic growth and proliferation, but little is known about its direct or indirect effects on cardiomyocytes. Cardiac muscle engineering is compromised by a particularly high rate of cardiomyocyte death. Here we hypothesized that PDGF-BB stimulation can (1) protect cardiomyocytes from apoptosis, (2) enhance myocyte content in and (3) consequently optimize contractile performance of engineered heart tissue (EHT). We investigated the effects of PDGF-receptor activation in neonatal rat heart monolayer- and EHT-cultures by isometric contraction experiments, cytomorphometry, (3)H-thymidine and (3)H-phenylalanine incorporation assays, quantitative PCR (calsequestrin 2, alpha-cardiac and skeletal actin, atrial natriuretic factor, alpha- and beta-myosin heavy chain), immunoblotting (activated caspase 3, Akt-phosphorylation), and ELISA (cell death detection). PDGF-BB did not induce hypertrophy or proliferation in cardiomyocytes, but enhanced contractile performance of EHT. This effect was concentration-dependent (E(max) 10 ng/ml) and maximal only after transient PDGF-BB stimulation (culture days 0-7; total culture duration: 12 days). Improvement of contractile function was associated with higher cardiomyocyte content, as a consequence of PDGF-BB mediated protection from apoptosis (lower caspase-3 activity particularly in cardiomyocytes in PDGF-BB treated vs. untreated EHTs). We confirmed the anti-apoptotic effect of PDGF-BB in monolayer cultures and observed that PI3-kinase inhibition with LY294002 attenuated PDGF-BB-mediated cardiomyocyte protection. We conclude that PDGF-BB does not induce hypertrophy or proliferation, but confers an anti-apoptotic effect on cardiomyocytes. Our findings suggest a further exploitation of PDGF-BB in cardiomyocyte protection in vivo and in vitro.


Assuntos
Apoptose , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Animais Recém-Nascidos , Becaplermina , Proliferação de Células , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Coração/fisiologia , Morfolinas/farmacologia , Fenilalanina/química , Proteínas Proto-Oncogênicas c-sis , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Engenharia Tecidual/métodos
13.
Cell Transplant ; 18(8): 847-53, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19520046

RESUMO

Bone marrow cells are used for cell therapy after myocardial infarction (MI) with promising results. However, cardiac persistence of transplanted cells is rather low. Here, we investigated strategies to increase the survival and cardiac persistence of mononuclear (MNC) and mesenchymal (MSC) bone marrow cells transplanted into infarcted rat hearts. MNC and MSC (male Fischer 344 rats) were treated with different doses of PDGF-BB prior to intramyocardial injection into border zone of MI (syngeneic females, permanent LAD ligation) and hearts were harvested after 5 days and 3 weeks. In additional experiments, untreated MNC and MSC were injected immediately after permanent or temporary LAD ligation and hearts were harvested after 48 h, 5 days, 3 weeks, and 6 weeks. DNA of the hearts was isolated and the number of donor cells was determined by quantitative real-time PCR with Y chromosome-specific primers. There was a remarkable though not statistically significant (p = 0.08) cell loss of approximately 46% between 5 days and 3 weeks in the control group, which was completely inhibited by treatment with high dose of PDGF-BB. Forty-eight hours after reperfusion only 10% of injected MSC or 1% for MNC were found in the heart, decreasing to 1% for MSC and 0.5% for MNC after 6 weeks. These numbers were lower than after permanent LAD ligation for both MNC and MSC at all time points studied. Treatment with PDGF-BB seems to prevent loss of transplanted bone marrow cells at later times presumably by inhibition of apoptosis, while reperfusion of the occluded artery enhances cell loss at early times putatively due to enhanced early wash-out. Further investigations are needed to substantially improve the persistence and survival of grafted bone marrow cells in infarcted rat hearts, in order to fully explore the therapeutic potential of this novel treatment modality for myocardial repair.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Transplante de Medula Óssea , Sobrevivência de Enxerto/efeitos dos fármacos , Leucócitos Mononucleares/efeitos dos fármacos , Infarto do Miocárdio/terapia , Reperfusão Miocárdica , Fator de Crescimento Derivado de Plaquetas/farmacologia , Indutores da Angiogênese/farmacologia , Indutores da Angiogênese/uso terapêutico , Animais , Becaplermina , Células da Medula Óssea/fisiologia , Transplante de Medula Óssea/métodos , Feminino , Leucócitos Mononucleares/fisiologia , Leucócitos Mononucleares/transplante , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Infarto do Miocárdio/fisiopatologia , Fator de Crescimento Derivado de Plaquetas/uso terapêutico , Proteínas Proto-Oncogênicas c-sis , Ratos , Ratos Endogâmicos F344 , Condicionamento Pré-Transplante/métodos
14.
Cardiovasc Res ; 81(4): 758-70, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19074160

RESUMO

AIMS: Moderate wine consumption is associated with a significant reduction of cardiovascular mortality. The molecular basis of this phenomenon remains unknown. Platelet-derived growth factor (PDGF) is an important contributor to atherogenesis. We investigated the effects of selected red and white wines on PDGF receptor (PDGFR) signalling in rat and human vascular smooth muscle cells (VSMCs). METHODS AND RESULTS: All red wines concentration dependently inhibited the ligand-induced tyrosine phosphorylation of the PDGFR, downstream signalling events such as mitogen activated protein (MAP) kinase activation (Erk 1/2) and induction of immediate early genes (Egr-1, c-fos), and PDGF-induced cellular responses, whereas all white wines had no effect. At concentrations achieved after wine consumption in humans, all red wines completely abolished PDGF-dependent VSMC proliferation and migration. Red wines also inhibited PDGFR phosphorylation in vascular tissue, and in human coronary smooth muscle cells. Quantitative analyses of all tested wines and of samples collected at various time points (Days 0-16) of the 'mash fermentation', which is only performed for red wine, revealed that flavonoids of the catechin family, which potently inhibit PDGFR signalling, are extracted from grape seeds and skins during this process and therefore accumulate specifically in red wine. The accumulation of flavonoids correlated with the inhibitory potency of red wines on PDGFR signalling. Furthermore, this procedure could be imitated by incubation of wines with shredded grape seeds, and flavonoid-enriched white wine inhibited the PDGFR as potently as red wines. CONCLUSION: Only red wines abrogate a critical pathogenic mechanism during atherogenesis, PDGFR signalling, in VSMCs. This effect is mediated by non-alcoholic constituents, which accumulate during the mash fermentation. Our findings offer a molecular explanation for the vasoprotective effects particularly of red wine. Therefore, future epidemiological studies should consider differential protective effects of red and white wine in vivo.


Assuntos
Fermentação , Flavonoides/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Fenóis/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Vinho , Animais , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Flavonoides/análise , Humanos , Masculino , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fenóis/análise , Fosforilação , Fator de Crescimento Derivado de Plaquetas/metabolismo , Polifenóis , Ratos , Ratos Endogâmicos WKY , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Fatores de Tempo , Vinho/análise
15.
J Biol Chem ; 283(12): 7864-76, 2008 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-18070887

RESUMO

Platelet-derived growth factor (PDGF) plays a critical role in the pathogenesis of proliferative diseases. NAD(P)H oxidase (Nox)-derived reactive oxygen species (ROS) are essential for signal transduction by growth factor receptors. Here we investigated the dependence of PDGF-AA-induced ROS production on the cytosolic Nox subunits Rac-1 and p47(phox), and we systematically evaluated the signal relay mechanisms by which the alphaPDGF receptor (alphaPDGFR) induces ROS liberation. Stimulation of the alphaPDGFR led to a time-dependent increase of intracellular ROS levels in fibroblasts. Pharmacological inhibitor experiments and enzyme activity assays disclosed Nox as the source of ROS. alphaPDGFR activation is rapidly followed by the translocation of p47(phox) and Rac-1 from the cytosol to the cell membrane. Experiments performed in p47(phox)(-/-) cells and inhibition of Rac-1 or overexpression of dominant-negative Rac revealed that these Nox subunits are required for PDGF-dependent Nox activation and ROS liberation. To evaluate the signaling pathway mediating PDGF-AA-dependent ROS production, we investigated Ph cells expressing mutant alphaPDGFRs that lack specific binding sites for alphaPDGFR-associated signaling molecules (Src, phosphatidylinositol 3-kinase (PI3K), phospholipase Cgamma, and SHP-2). Lack of PI3K signaling (but not Src, phospholipase Cgamma, or SHP-2) completely abolished PDGF-dependent p47(phox) and Rac-1 translocation, increase of Nox activity, and ROS production. Conversely, a mutant alphaPDGFR able to activate only PI3K was sufficient to mediate these subcellular events. Furthermore, the catalytic PI3K subunit p110alpha (but not p110beta) was identified as the crucial isoform that elicits alphaPDGFR-mediated production of ROS. Finally, bromodeoxyuridine incorporation and chemotaxis assays revealed that the lack of ROS liberation blunted PDGF-AA-dependent chemotaxis but not cell cycle progression. We conclude that PI3K/p110alpha mediates growth factor-dependent ROS production by recruiting p47(phox) and Rac-1 to the cell membrane, thereby assembling the active Nox complex. ROS are required for PDGF-AA-dependent chemotaxis but not proliferation.


Assuntos
Membrana Celular/metabolismo , Fibroblastos/metabolismo , NADPH Oxidases/metabolismo , Neuropeptídeos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Classe I de Fosfatidilinositol 3-Quinases , Citoplasma/metabolismo , Fibroblastos/citologia , Humanos , Camundongos , Camundongos Knockout , NADPH Oxidases/genética , Neuropeptídeos/genética , Fosfatidilinositol 3-Quinases/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/agonistas , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fatores de Tempo , Proteínas rac de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/genética
16.
FEBS Lett ; 580(30): 6769-76, 2006 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-17141222

RESUMO

Regulation of growth factor dependent cell survival is crucial for development and disease progression. Here, we report a novel function of Src kinases as a negative regulator of platelet-derived growth factor (PDGF) dependent cell survival. We characterized a series of PDGF alpha receptor (PDGFRA) mutants, which lack the binding sites for Src, phosphatidylinositol 3'-kinase (PI3K), SHP-2 or phospholipase C-gamma. We found that PDGFRA-dependent cell survival was mainly mediated through activation of PI3K, and was negatively regulated by Src. Characterization of the downstream signaling events revealed that PI3K activates the protein kinase Akt, which in turn phosphorylates and thus inactivates proapoptotic Forkhead transcription factors. Src phosphorylates the ubiquitin-ligase c-Cbl, which is required for degradation of the activated receptor. Consequently, overexpression of c-Cbl prevented PDGFRA-mediated cell survival, whereas it did not affect this response, when Src was unable to associate with the receptor. This novel function of Src in antiapoptotic signaling introduces Src kinases as an interesting therapeutic target in apoptosis related diseases.


Assuntos
Apoptose , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Quinases da Família src/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular , Regulação para Baixo , Ativação Enzimática , Fatores de Transcrição Forkhead/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Mutação/genética , Fosforilação , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética
17.
Cardiovasc Res ; 71(2): 331-41, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16545786

RESUMO

OBJECTIVE: Reactive oxygen species (ROS) produced by NAD(P)H oxidases (Nox) play a significant role in the pathophysiology of cardiovascular diseases. Expression and activity of NAD(P)H oxidases are regulated by growth factors such as angiotensin II and platelet-derived growth factor (PDGF). We characterized the effects of the novel Nox inhibitor VAS2870 on PDGF-dependent ROS liberation and cellular events in vascular smooth muscle cells (VSMC). METHODS AND RESULTS: PDGF-BB increased NAD(P)H oxidase activity (lucigenin-enhanced chemiluminescence) and intracellular ROS levels (detected by confocal laserscanning microscopy using 2,7-DCF) to 229+/-9% and 362+/-54% at 1 and 2 h, respectively. Preincubation with VAS2870 (10 and 20 microM) completely abolished PDGF-mediated NAD(P)H oxidase activation and ROS production. Since ROS are involved in various growth factor-induced cellular functions, the influence of VAS2870 on PDGF-induced DNA synthesis and chemotaxis was determined. PDGF promoted a 4.2+/-0.2-fold increase of VSMC migration (modified Boyden chamber, p<0.01) and increased DNA synthesis by maximally 3.2+/-0.4-fold (BrdU incorporation, p<0.01) in a concentration-dependent manner. Preincubation with VAS2870 (0.1-20 microM) did not affect PDGF-induced cell cycle progression. However, it abolished PDGF-dependent chemotaxis of VSMC in a concentration-dependent manner (100% inhibition at 10 microM). These findings were related to PDGF-dependent signaling events. Western blot analyses using phospho-specific antibodies revealed that the downstream signaling molecules Akt, Erk, and Src were activated by PDGF. However, VAS2870 blocked PDGF-dependent activation of Src, but not of Akt and Erk, in a concentration-dependent manner. CONCLUSIONS: VAS2870 effectively suppresses growth factor-mediated ROS liberation in VSMC. Furthermore, it completely inhibits PDGF-dependent VSMC migration, whereas it does not affect DNA synthesis. These divergent effects reflect the critical role of Src activity, which-in contrast to Akt and Erk-appears to be redox-sensitive and is absolutely required for PDGF-induced chemotaxis, but not cell cycle progression.


Assuntos
Benzoxazóis/farmacologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , NADPH Oxidases/antagonistas & inibidores , Fator de Crescimento Derivado de Plaquetas/metabolismo , Triazóis/farmacologia , Animais , Aorta Torácica , Becaplermina , Western Blotting/métodos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , DNA/biossíntese , Inibidores Enzimáticos/farmacologia , Imunoprecipitação , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-sis , Ratos , Ratos Endogâmicos WKY , Espécies Reativas de Oxigênio/análise , Espécies Reativas de Oxigênio/metabolismo , Quinases da Família src/metabolismo
18.
J Biol Chem ; 280(14): 14168-76, 2005 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-15640155

RESUMO

Peptide growth factors contribute to the pathogenesis of cardiovascular diseases by inducing a variety of cellular responses including anti-apoptotic effects. Several of the signaling molecules that are activated by growth factor receptors such as Src family kinases (Src), phosphatidylinositol 3'-kinase (PI3K), phospholipase Cgamma (PLCgamma), Ras, and SHP-2 were shown to mediate survival signals. We systematically investigated the relative contribution of each signaling molecule for growth factor-dependent cell survival in vascular smooth muscle cells (VSMC). Our approach was the use of mutated plateletderived growth factor (PDGF) beta-receptors (betaPDGFR) in which the tyrosine residues required for binding of each signaling molecule were individually mutated to phenylalanine. To bypass endogenous PDGFR in VSMC we used chimeric receptors (ChiRs), containing the extracellular domain of the macrophage colony-stimulating factor (M-CSF) receptor and the cytoplasmic domain of the wild type (WT) or mutated betaPDGFR. Selective activation of the ChiR-WT with M-CSF significantly reduced apoptosis to the same extent as PDGF-BB in non-transfected cells. Deletion of the binding site for PI3K, but not for Src, RasGAP, SHP-2, or PLCgamma, completely abolished the anti-apoptotic effect. Consistently, a ChiR mutant that only binds PI3K was fully able to mediate cell survival as efficiently as the ChiR-WT. Furthermore, the PDGF-dependent anti-apoptotic effect in non-transfected cells was completely abolished by the PI3K inhibitor wortmannin, whereas inhibitors of Src, PLCgamma, ERK, or p38 MAP kinase had no effect. The exploration of downstream signaling events revealed that PDGF-BB activates the anti-apoptotic Akt signaling pathway in a PI3K-dependent manner. Moreover, Akt phosphorylates and thus inactivates the pro-apoptotic proteins BAD and Forkhead transcription factors (FKHR, FKHRL1). We conclude that growth factor-dependent cell survival in VSMC is mediated only by activation of the PI3K/Akt pathway, whereas all other receptor-associated signaling molecules do not play a significant role.


Assuntos
Apoptose/fisiologia , Substâncias de Crescimento/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/fisiologia , Animais , Becaplermina , Proteínas de Transporte/metabolismo , Sobrevivência Celular , Células Cultivadas , Ativação Enzimática , Fatores de Transcrição Forkhead , Humanos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Proteínas Nucleares/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-sis , Ratos , Ratos Wistar , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/metabolismo , Proteína de Morte Celular Associada a bcl
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA