Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Cell Cardiol ; 43(2): 137-47, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17583725

RESUMO

Mitogen-activated protein (MAP) kinases have been implicated in hemodynamic load induced heart failure. Both angiotensin II (Ang II) and mechanical stretch activate MAP kinases in cardiac myocytes. In this study, we used a neonatal rat ventricular myocyte (NRVM) model to determine the role of focal-adhesion kinase (FAK) in beta1 integrin mediated MAP kinase activation in response to mechanical stretch in presence and absence of Ang II receptor blockade (ATB). NRVM plated on deformable membranes coated with collagen IV were exposed to 20% equiaxial static-stretch. beta1 integrin signaling was blocked by adenovirus-mediated expression of a dominant-negative form of beta1D integrin (tac-beta1D). FAK signaling was disrupted by infecting NRVM with adenovirus expressing FAK-related non-kinase (FRNK). Western blot analysis was used to assess the phosphorylation of MAP kinases. In the presence and absence of ATB, mechanical stretch caused maximal phosphorylation of ERK, p38 and JNK at 5 min, which was significantly attenuated in NRVM expressing tac-beta1D. In the presence of ATB, FRNK overexpression significantly increased basal phosphorylation of ERK (40.2+/-8.6% P<0.05), p38 (39.5+/-11.7%, P<0.05), JNK (86+/-29.4%, P<0.05) and stretch-induced p38 (48.1+/-8.7%, P<0.05) and JNK (85.0+/-19.4%, P<0.05) phosphorylation. However, in the absence of ATB, FRNK overexpression significantly reduced basal and stretch-induced phosphorylation of only ERK. Examination of FAK activation revealed that beta1 integrin was required for stretch-induced phosphorylation of FAK at Y397 and Y925, but not Y861. In summary, mechanical stretch-activated ERK1/2, p38 and JNK through FAK independent and dependent mechanisms. Beta1 integrin was required for FAK independent activation of all three MAP kinases, whereas cross-talk between beta1 integrin and Ang II receptors mediated FAK dependent regulation of ERK1/2.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Integrina beta1/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miócitos Cardíacos/enzimologia , Animais , Animais Recém-Nascidos , Ativação Enzimática , Fosforilação , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Estresse Mecânico , Fatores de Tempo , Tirosina/metabolismo
3.
Circ Res ; 88(3): 298-304, 2001 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-11179197

RESUMO

Ventricular pacing leads to a dilated myopathy in which cell death and myocyte hypertrophy predominate. Because angiotensin II (Ang II) stimulates myocyte growth and triggers apoptosis, we tested whether canine myocytes express the components of the renin-angiotensin system (RAS) and whether the local RAS is upregulated with heart failure. p53 modulates transcription of angiotensinogen (Aogen) and AT(1) receptors in myocytes, raising the possibility that enhanced p53 function in the decompensated heart potentiates Ang II synthesis and Ang II-mediated responses. Therefore, the presence of mRNA transcripts for Aogen, renin, angiotensin-converting enzyme, chymase, and AT(1) and AT(2) receptors was evaluated by reverse transcriptase-polymerase chain reaction in myocytes. Changes in the protein expression of these genes were then determined by Western blot in myocytes from control dogs and dogs affected by congestive heart failure. p53 binding to the promoter of Aogen and AT(1) receptor was also determined. Ang II in myocytes was measured by ELISA and by immunocytochemistry and confocal microscopy. Myocytes expressed mRNAs for all the constituents of RAS, and heart failure was characterized by increased p53 DNA binding to Aogen and AT(1). Additionally, protein levels of Aogen, renin, cathepsin D, angiotensin-converting enzyme, and AT(1) were markedly increased in paced myocytes. Conversely, chymase and AT(2) proteins were not altered. Ang II quantity and labeling of myocytes increased significantly with cardiac decompensation. In conclusion, dog myocytes synthesize Ang II, and activation of p53 function with ventricular pacing upregulates the myocyte RAS and the generation and secretion of Ang II. Ang II may promote myocyte growth and death, contributing to the development of heart failure.


Assuntos
Insuficiência Cardíaca/fisiopatologia , Sistema Renina-Angiotensina/fisiologia , Função Ventricular , Actinas/metabolismo , Angiotensina II/metabolismo , Animais , Ligação Competitiva , Western Blotting , Estimulação Cardíaca Artificial , Catepsina D/metabolismo , Quimases , Cães , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Imuno-Histoquímica , Microscopia Confocal , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Angiotensina , Receptor Tipo 2 de Angiotensina , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Renina/genética , Renina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
4.
Neth Heart J ; 9(2): 65-67, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-25696696
5.
Regul Pept ; 91(1-3): 1-11, 2000 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-10967197

RESUMO

Angiotensin II, the effector peptide of the renin-angiotensin system, has been demonstrated to be involved in the regulation of cellular growth of several tissues in response to developmental, physiological, and pathological processes. The recent identification of renin-angiotensin system components and localization of angiotensin II receptors in cardiac tissue suggests that locally synthesized Ang II can modulate functional and growth responses in cardiac tissue. In this review, regulation of the cardiac RAS is discussed, with an emphasis on growth-related Ang II signal transduction systems.


Assuntos
Coração/fisiologia , Sistema Renina-Angiotensina/fisiologia , Aldosterona/metabolismo , Angiotensina II/metabolismo , Angiotensinas/fisiologia , Animais , Animais Geneticamente Modificados/fisiologia , Células Cultivadas , Humanos , Modelos Biológicos , Miocárdio/citologia , Miocárdio/metabolismo , Renina/fisiologia , Transdução de Sinais
6.
Hypertension ; 35(6): 1191-6, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10856262

RESUMO

-Cardiotrophin-1, an interleukin-6-related cytokine, stimulates the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway and induces cardiac myocyte hypertrophy. In this study, we demonstrate that cardiotrophin-1 induces cardiac myocyte hypertrophy in part by upregulation of a local renin-angiotensin system through the JAK/STAT pathway. We found that cardiotrophin-1 increased angiotensinogen mRNA expression in cardiac myocytes via STAT3 activation. Tyrosine phosphorylation of STAT3 by cardiotrophin-1 treatment resulted in STAT3 homodimer binding to the St-domain in the angiotensinogen gene promoter, which lead to promoter activation in a transient transfection assay. Cardiotrophin-1-induced STAT3 tyrosine phosphorylation and binding to the St-domain were suppressed by AG490, a specific JAK2 inhibitor, which also attenuated cardiotrophin-1-stimulated angiotensinogen promoter activity. Cardiotrophin-1 did not activate the angiotensinogen gene promoter that contained a substitution mutation within the St-domain. Finally, losartan, an angiotensin II type 1 receptor antagonist, significantly attenuated cardiotrophin-1-induced hypertrophy of neonatal rat cardiac myocytes. Angiotensin II is known to induce cardiac myocyte hypertrophy by activating the G-protein-coupled angiotensin II type 1 receptor. Our results suggest that upregulation of angiotensinogen and angiotensin II production contribute to cardiotrophin-1-induced cardiac myocyte hypertrophy and emphasize an important interaction between G-protein-coupled and cytokine receptors.


Assuntos
Angiotensinogênio/genética , Citocinas/fisiologia , Proteínas de Ligação a DNA/fisiologia , Miocárdio/metabolismo , RNA Mensageiro/metabolismo , Transativadores/fisiologia , Antagonistas de Receptores de Angiotensina , Animais , Comunicação Autócrina , Cardiomegalia/etiologia , Cardiomegalia/prevenção & controle , Citocinas/antagonistas & inibidores , Miocárdio/citologia , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/fisiologia , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina , Receptor Tipo 2 de Angiotensina , Fator de Transcrição STAT3 , Transativadores/metabolismo , Tirosina/metabolismo
7.
Basic Res Cardiol ; 95(6): 485-90, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11192370

RESUMO

We previously demonstrated the presence of components for a renin-angiotensin system in fibroblasts cultured from neonatal rat ventricles, the regulation of expression of which has not been studied. Since glucocorticoids and beta-adrenergic stimuli have been implicated in cardiac hypertrophy, and function as regulators of the circulating renin-angiotensin system, we examined the effects of dexamethasone and isoproterenol on angiotensinogen mRNA levels and protein secretion in cultured neonatal rat cardiac fibroblasts. Treatment of cardiac fibroblasts for 8 h with 10 micromol/l isoproterenol or 100 nmol/l dexamethasone increased angiotensinogen mRNA levels by 246 +/- 7% and 1406 +/- 207%, respectively. Over 24 h, dexamethasone and isoproterenol increased angiotensinogen secretion by 148 +/- 32% and 123 +/- 26%, respectively. Angiotensin II, which has been reported to be a positive regulator of angiotensinogen synthesis and secretion in liver, markedly attenuated the effects of dexamethasone and isoproterenol on angiotensinogen mRNA expression and secretion. In the presence of 1 micromol/l angiotensin II, the stimulation in angiotensinogen secretion observed with dexamethasone and isoproterenol was decreased by 62% and 76%, respectively. The negative feedback of angiotensin II on angiotensinogen expression was primarily mediated through the type one angiotensin II (AT1) receptor (IC50 = 0.30 +/- 0.02 nmol/l). In summary, results from this study demonstrate that angiotensinogen mRNA levels and protein secretion in cardiac fibroblasts are positively regulated by glucocorticoid and beta-adrenergic stimulation. In addition, angiotensinogen production by cardiac fibroblasts is under negative feedback control of angiotensin II.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Angiotensinogênio/genética , Dexametasona/farmacologia , Fibroblastos/fisiologia , Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Isoproterenol/farmacologia , Miocárdio/citologia , Angiotensina II/antagonistas & inibidores , Animais , Animais Recém-Nascidos/fisiologia , Técnicas de Cultura , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
8.
Circ Res ; 85(7): 643-50, 1999 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-10506489

RESUMO

Angiotensin II, the effector peptide of the renin-angiotensin system, regulates cellular growth in response to developmental, physiological, and pathological processes. The identification of renin-angiotensin system components and angiotensin II receptors in cardiac tissue suggests the existence of an autocrine/paracrine system that has effects independent of angiotensin II derived from the circulatory system. To be functional, a local renin-angiotensin system should produce sufficient amounts of the autocrine and/or paracrine factor to elicit biological responses, contain the final effector (angiotensin II receptor), and respond to humoral, neural, and/or mechanical stimuli. In this review, we discuss evidence for a functional cardiac renin-angiotensin system.


Assuntos
Coração/fisiologia , Sistema Renina-Angiotensina/fisiologia , Angiotensina II/biossíntese , Angiotensinogênio/metabolismo , Animais , Animais Geneticamente Modificados/fisiologia , Baixo Débito Cardíaco/metabolismo , Baixo Débito Cardíaco/fisiopatologia , Humanos , Miocárdio/citologia , Miocárdio/metabolismo , Peptidil Dipeptidase A/metabolismo , Renina/metabolismo
9.
Am J Cardiol ; 83(12A): 44H-47H, 1999 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-10750586

RESUMO

Conditioned medium of cardiac fibroblasts was found to induce protein synthesis and signal transduction events rapidly, and to increase angiotensinogen messenger RNA (mRNA) levels in neonatal rat ventricular myocytes. Within 4 hours, fibroblast-conditioned medium (FCM) stimulated protein synthesis in cardiac myocytes, independent of the contractile state, and induced marked increases within 24 hours in total protein content. Endothelin- released by cardiac fibroblasts was not responsible for the stimulation of protein synthesis. FCM rapidly activated signal transduction events in cardiac myocytes associated with hypertrophic stimuli, including: (1) increased tyrosine phosphorylation of several prominent protein bands; (2) mitogen-activated protein kinases (ERK 1 and ERK 2); and (3) protein kinase C. Finally, FCM caused an increase at 8 hours in angiotensinogen mRNA levels of cardiac myocytes, whereas no effect was observed on mRNA levels for renin or the type 1 angiotensin II receptor (AT1). Our results suggest that cardiac fibroblasts produce a factor that rapidly activates cardiac myocyte growth through a membrane receptor that couples to conventional signal transduction pathways.


Assuntos
Fibroblastos/metabolismo , Miocárdio/metabolismo , Comunicação Parácrina/fisiologia , Sistema Renina-Angiotensina/fisiologia , Angiotensinogênio/genética , Angiotensinogênio/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Meios de Cultivo Condicionados , Endotelina-1/metabolismo , Fibroblastos/citologia , Coração/crescimento & desenvolvimento , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miocárdio/citologia , Fosforilação , Fosfotransferases/metabolismo , Proteína Quinase C/metabolismo , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia
11.
Hypertension ; 30(6): 1362-8, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9403554

RESUMO

The spontaneously hypertensive rat (SHR) exhibits a transition from stable compensated left ventricular (LV) hypertrophy to heart failure (HF) at a mean age of 21 months that is characterized by a decrease in alpha-myosin heavy chain (alpha-MHC) gene expression and increases in the expression of the atrial natriuretic factor (ANF), pro-alpha1(III) collagen, and transforming growth factor beta1 (TGF-beta1) genes. We tested the hypotheses that angiotensin-converting enzyme inhibition (ACEI) in SHR would prevent and reverse HF-associated changes in gene expression when administered prior to and after the onset of HF, respectively. We also investigated the effect of ACEI on circulating and cardiac components of the renin-angiotensin system. ACEI (captopril 2 g/L in the drinking water) was initiated at 12, 18, and 21 months of age in SHR without HF and in SHR with HF. Results were compared with those of age-matched normotensive Wistar-Kyoto (WKY) rats, and to untreated SHR with and without evidence of HF. ACEI initiated prior to failure prevented the changes in alpha-MHC, ANF, pro-alpha1(III) collagen, and TGF-beta1 gene expression that are associated with the transition to HF. ACEI initiated after the onset of HF lowered levels of TGF-beta1 mRNA by 50% (P<.05) and elevated levels of alpha-MHC mRNA two- to threefold (P<.05). Circulating levels of renin and angiotensin I were elevated four- to sixfold by ACEI, but surprisingly, plasma levels of angiotensin II were not reduced. ACEI increased LV renin mRNA levels in WKY and SHR by two- to threefold but did not influence LV levels of angiotensinogen mRNA. The results suggest that the anti-HF benefits of ACEI in SHR may be mediated, at least in part, by effects on the expression of specific genes, including those encoding alpha-MHC, ANF, TGF-beta1, pro-alpha1(III) collagen, and renin-angiotensin system components.


Assuntos
Envelhecimento/fisiologia , Captopril/farmacologia , Cardiomegalia/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Insuficiência Cardíaca/fisiopatologia , Coração/fisiopatologia , Hipertensão/fisiopatologia , Renina/biossíntese , Análise de Variância , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Angiotensinogênio/biossíntese , Animais , Fator Natriurético Atrial/biossíntese , Cardiomegalia/fisiopatologia , Coração/crescimento & desenvolvimento , Coração/fisiologia , Insuficiência Cardíaca/metabolismo , Hipertensão/metabolismo , Masculino , Cadeias Pesadas de Miosina/biossíntese , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Transcrição Gênica , Fator de Crescimento Transformador beta/biossíntese
12.
J Mol Cell Cardiol ; 29(9): 2513-24, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9299374

RESUMO

The octapeptide, angiotensin II, has a modulatory role on cardiac cellular growth associated with hypertension and in compensatory remodeling following myocardial infarction. The molecular signal transduction pathways that participate in these and other cellular actions in response to angiotensin II are presently being elucidated. The signal transducers and activators of transcription (STAT) pathway directly links cytokine and growth factor receptors with transcriptional activity. We provide evidence that the G protein-linked, angiotensin II, AT1-receptor couples to activation of the STAT pathway in neonatal rat cardiac myocytes. Angiotensin II induces primarily sis-inducing factor (SIF) B and to a lesser extent SIF-C and SIF-A. The EC50 of this response was 40 nM and Stat1 and Stat3 proteins were identified as components of the SIF complexes. Stat1 and Stat3 were tyrosine phosphorylated five-fold and three-fold, respectively, over control levels following angiotensin II treatment of cardiac myocytes. Phosphorylation of Stat1 and Stat3 proteins was rapid (5 min) and sustained (60 min). Jak2 was also tyrosine phosphorylated eight-fold by angiotensin II treatment, and phosphorylated Stat1 and Stat3 proteins co-immunoprecipitated with activated Jak2 kinase. Selective inhibition of Jak2 kinase with AG-490 blocked formation of angiotensin II induced SIF complexes, suggesting that Jak2 kinase is required for cardiomyocyte SIF induction. In addition, Jak2, Stat1 and Stat3 proteins co-immunoprecipitated with the AT1-receptor. These are the first data to demonstrate coupling of a G-protein coupled receptor, AT1, to the JAK-STAT pathway in primary cultured cardiac myocytes and suggest that this pathway may be involved in transcriptional regulation by angiotensin II.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Miocárdio/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Receptores de Angiotensina/metabolismo , Transativadores/metabolismo , Tirfostinas , Angiotensina II/farmacologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Proteínas de Ligação a DNA/efeitos dos fármacos , Coração/efeitos dos fármacos , Imidazóis/farmacologia , Janus Quinase 2 , Losartan , Miocárdio/citologia , Nitrilas/farmacologia , Fosforilação , Proteínas Tirosina Quinases/efeitos dos fármacos , Ratos , Receptor Tipo 1 de Angiotensina , Receptor Tipo 2 de Angiotensina , Receptores de Angiotensina/efeitos dos fármacos , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Tetrazóis/farmacologia , Transativadores/efeitos dos fármacos , Transcrição Gênica , Tirosina/metabolismo
13.
J Mol Cell Cardiol ; 29(11): 2893-902, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9405164

RESUMO

Angiotensin II (Ang II), the effector peptide of the renin-angiotensin system (RAS), regulates volume and electrolyte homeostasis and is involved in cardiac and vascular cellular growth in humans and other species. This system, which has been conserved throughout evolution, plays an important role in cardiac and vascular pathology associated with hypertension, coronary heart disease, myocarditis and congestive heart failure. The traditional RAS is viewed as a system in which circulating Ang II is delivered to target organs and cells. However, in the past decade, a local RAS has been described in cardiac cells, providing evidence for autocrine and paracrine pathways by which biological actions of Ang II could be mediated. The critical actions of Ang II are mediated primarily through the AT1, G-protein (guanylyl nucleotide binding protein) coupled receptor. In addition to coupling to conventional G-protein signal transduction pathways, the AT1 receptor was recently shown to increase the tyrosine phosphorylation of several intracellular substrates, including the STAT (Signal Transducers and Activators of Transcription) family of novel transcription factors, in rat cardiac fibroblasts, myocytes and vascular smooth muscle cells, and AT1 receptor transfected CHO cells. It has been shown that Ang II stimulates the tyrosine phosphorylation and nuclear translocation of Stat1 (Stat 91) and Stat3 (Stat 92). Angiotensin II acting directly through the AT1 receptor, induces the formation of a complex of STAT proteins termed SIF (sis-inducing factor) which binds the DNA sequence, SIE (sis-inducing element) present in the promotor element of many genes. This provides evidence for a direct role of Ang II in mediating inflammatory and remodeling responses through the JAK-STAT pathway. Thus, it is likely that the JAK-STAT pathway has an important role in Ang II-mediated effects on gene transcription, cardiac and vascular cellular growth/development, and inflammatory responses.


Assuntos
Angiotensina II/fisiologia , Coração/fisiologia , Proteínas Tirosina Quinases/fisiologia , Transdução de Sinais/fisiologia , Transativadores/fisiologia , Animais , Divisão Celular/fisiologia , Conformação Proteica , Receptor Tipo 1 de Angiotensina , Receptor Tipo 2 de Angiotensina , Receptores de Angiotensina/fisiologia
14.
Mol Cell Biochem ; 157(1-2): 15-21, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8739224

RESUMO

Angiotensin II has been demonstrated to be involved in the regulation of cellular growth of several tissues in response to developmental, physiological, and pathophysiological processes. Angiotensin II has been implicated in the developmental growth of the left ventricle in the neonate and remodeling of the heart following chronic hypertension and myocardial infarction. The inhibition of DNA synthesis and collagen deposition in myocardial interstitium following myocardial infarction by angiotensin converting enzyme inhibitor, suggests that angiotensin II mediates interstitial and perivascular fibrobrosis by preventing fibroblast proliferation. In the past, little attention was focused on the identity and functional roles of cardiac fibroblasts. Recent in vitro studies utilizing cultured cardiac fibroblasts demonstrate that angiotensin II, acting via the AT1 receptor, initiates intracellular signalling pathways in common with those of peptide growth factors. Below, we describe growth-related aspects of cardiac fibroblasts with respect to angiotensin II receptors, conventional and novel signal transduction systems, secretion of extracellular matrix proteins and growth factors, and localization of renin-angiotensin system components.


Assuntos
Angiotensina II/fisiologia , Coração/fisiologia , Transdução de Sinais , Angiotensina II/farmacologia , Animais , Citocinas/fisiologia , Matriz Extracelular/fisiologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Substâncias de Crescimento/fisiologia , Coração/efeitos dos fármacos , Humanos , Hipertensão/fisiopatologia , Recém-Nascido , Infarto do Miocárdio/fisiopatologia , Miocárdio/citologia , Miocárdio/metabolismo , Proteína Quinase C/metabolismo
15.
Am J Physiol ; 269(5 Pt 2): H1791-802, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7503279

RESUMO

To date, the demonstration that the molecular components of the renin-angiotensin system (RAS) are present in adult ventricular myocytes is lacking. In addition, whether the RAS is upregulated under conditions of overload and myocyte hypertrophy in vivo remains to be determined. By employing an in vivo model of ischemic cardiomyopathy in rats, we document that adult myocytes express genes for renin, angiotensinogen, angiotensin-converting enzyme (ACE), and angiotensin II (ANG II) receptors. Moreover, renin, ACE, and ANG II receptor mRNAs increased in stressed myocytes undergoing cellular hypertrophy. At the protein level, the percentage of myocytes containing renin, ANG I, and ANG II was significantly increased in the overloaded heart. The number of binding sites for ANG II per myocyte also markedly increased under this setting. These results provide direct evidence of the existence of a myocyte RAS, which may be activated in pathological states of the heart to support myocyte growth and contractile function.


Assuntos
Miocárdio/metabolismo , Sistema Renina-Angiotensina/fisiologia , Angiotensinogênio/metabolismo , Angiotensinas/metabolismo , Animais , Sequência de Bases , Coração/fisiopatologia , Ventrículos do Coração , Imuno-Histoquímica , Masculino , Sondas Moleculares , Dados de Sequência Molecular , Miocárdio/citologia , Peptidil Dipeptidase A/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Angiotensina/metabolismo , Renina/metabolismo
16.
Biotechniques ; 18(6): 1014-20, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7546701

RESUMO

We have developed a sensitive, high-throughput bioassay to quantify angiotensin II in culture medium. Using Chinese hamster ovary cells that stably express a transfected angiotensin II receptor as target cells, we demonstrated that an agonist-stimulated myelin basic protein kinase response can be used as a basis of quantitative bioassay for angiotensin II. The assay permits detection of as little as 10 pg of angiotensin II in 1 mL of medium and offers an excellent alternative to HPLC and radioimmunoassay. This approach may also be applicable for quantification of other peptide hormones or growth factors produced by cell cultures.


Assuntos
Angiotensina II/análise , Bioensaio/métodos , Antagonistas de Receptores de Angiotensina , Animais , Compostos de Bifenilo , Células CHO/enzimologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Cricetinae , Meios de Cultura , Meios de Cultura Livres de Soro , Quinase 3 da Glicogênio Sintase , Imidazóis , Losartan , Ratos , Receptores de Angiotensina/genética , Sensibilidade e Especificidade , Tetrazóis , Transfecção
17.
J Mol Cell Cardiol ; 27(5): 1151-60, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7473773

RESUMO

We have previously shown that angiotensin II (AII) is a mitogen for neonatal rat cardiac fibroblasts. However, the signaling events that lead to fibroblast cell growth in response to AII remain to be elucidated. Mitogen-activated protein (MAP) kinases are cytosolic serine/threonine kinases which have been shown to be activated in quiescent cells by diverse growth stimuli, thereby being linked to growth regulatory pathways. This study was designed to determine whether MAP-kinase activation occurred in response to AII/receptor coupling in neonatal rat cardiac fibroblasts and the role of MAP-kinase activation in the AII-induced proliferation of these cells. Immunoblot analysis of MAP-kinase isoforms revealed predominantly p44 with less p42 MAP-kinase in rat cardiac fibroblasts. Both isoforms were activated upon stimulation of the cells with AII for 5 min or platelet derived growth factor-BB for 10 min. Angiotensin II stimulated MAP-kinase in a dose-dependent fashion with an EC50 of 2.5 nM. Two minutes following stimulation with 1 microM AII MAP-kinase activity increased from 90 +/- 17.9 to 477.5 +/- 75.9 pmol/min/mg protein, P < 0.05, n = 4. A smaller, sustained, secondary increase in MAP-kinase activity from 37.7 +/- 5.3 to 110.9 +/- 15.3 pmol/min/mg protein, P < 0.05, n = 4, was observed in response to AII between 120-150 minutes following receptor occupancy. The responses to AII were markedly attenuated by the AT1 receptor antagonist EXP3174. Stimulation of the cells with carbachol induced the first but not the second phase of MAP-kinase activity and this compound had no effect on cellular growth. The second phase of MAP-kinase activity 2-2.5 h after AII stimulation, paralleled data demonstrating that a 2-3 h receptor occupancy with AII was necessary to induce DNA synthesis and fibroblast proliferation. These results indicate that AII stimulates a biphasic activation of MAP-kinase by the AT1 receptor and that this pathway may participate in the AII induced mitogenic response in cardiac fibroblasts.


Assuntos
Angiotensina II/análogos & derivados , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Miocárdio/enzimologia , Angiotensina II/metabolismo , Angiotensina II/farmacologia , Antagonistas de Receptores de Angiotensina , Animais , Animais Recém-Nascidos , Becaplermina , Carbacol/farmacologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , DNA/biossíntese , Ativação Enzimática/efeitos dos fármacos , Fibroblastos , Ventrículos do Coração , Imidazóis/farmacologia , Immunoblotting , Losartan , Miocárdio/citologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-sis , Ratos , Ratos Sprague-Dawley , Receptores de Angiotensina/metabolismo , Tetrazóis/farmacologia
18.
Mol Cell Biochem ; 146(1): 79-89, 1995 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-7651382

RESUMO

The octapeptide angiotensin II mediates the physiological actions of the renin-angiotensin system through activation of several angiotensin II receptor subtypes; in particular the AT1. In many tissues, the presence of multiple angiotensin II receptor subtypes, together with a low number of receptors, makes it difficult to study biological responses to physiological concentrations (10(-11)-10(-9) M) of angiotensin II. Also, cultured cells show diminished angiotensin II receptor binding with respect to time in culture and passage number. To address these problems, we expressed the recombinant AT1A receptor in CHO-K1 cells. The stably transfected receptor was characterized using radioligand binding studies and functional coupling to cytosolic free calcium. Radioligand binding of [125I] angiotensin II to the angiotensin II receptor was specific, saturable, reversible and modulated by guanine nucleotides. Like the endogenous AT1A receptor, reported in a variety of tissues, the specific, noncompetitive, nonpeptide AII receptor antagonist, EXP3174, blocked binding of [125I] angiotensin II to the transfected receptor. Scatchard analysis demonstrated that the transfected receptor had a dissociation constant of 1.9 nM with a density of 3.4 pmol/mg protein. An important feature of many of the responses to angiotensin II is the rapid desensitization that occurs following agonist occupancy and the development of tachyphylaxis. In AT1A receptor transfected CHO-K1 cells, angiotensin II (10(-9) M) stimulated a rapid increase in cytosolic free calcium that was completely desensitized within 50 sec following receptor occupancy. Agonist induced desensitization was unaffected when receptor internalization was blocked by pretreatment with concanavalin A or incubation at 4 degrees C, and no changes in AT1A receptor affinity or number were observed. Receptor desensitization was also unaffected by inhibition or activation of protein kinase C. Thus, we have established a permanent, high-level transfectant of the AT1A receptor in CHO-K1 cells and have shown that these receptors rapidly desensitize following exposure to physiological concentrations of agonist. The mechanism of rapid desensitization is not related to receptor sequestration, internalization or controlled by PKC phosphorylation. This provides an excellent model for studying AII actions mediated through a specific receptor subtype, at subnanomolar concentrations.


Assuntos
Angiotensina II/farmacologia , Receptores de Angiotensina/genética , Angiotensina II/metabolismo , Angiotensina III/farmacologia , Antagonistas de Receptores de Angiotensina , Animais , Anti-Hipertensivos/farmacologia , Sequência de Bases , Células CHO , Cálcio/metabolismo , Cálcio/farmacologia , Membrana Celular/metabolismo , Concanavalina A/farmacologia , Cricetinae , Expressão Gênica/efeitos dos fármacos , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/farmacologia , Imidazóis/farmacologia , Losartan , Potenciais da Membrana/efeitos dos fármacos , Dados de Sequência Molecular , Piridinas/farmacologia , Ratos , Receptores de Angiotensina/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Tetrazóis/farmacologia , Transfecção , Verapamil/farmacologia
19.
Anal Biochem ; 223(2): 239-50, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7887470

RESUMO

We have developed a multiplex, competitive, reverse-transcriptase polymerase chain reaction (RT-PCR) method which measures absolute levels of renin, angiotensinogen, and the housekeeping transcript elongation factor-1 alpha (EF-1 alpha) mRNA. Sample RNA was simultaneously titrated with serial dilutions of renin, angiotensinogen, and EF-1 alpha competitor RNAs which flanked the endogenous concentrations of target transcripts. The samples were coreverse transcribed in the presence of random primers and resulting first-strand cDNA was coamplified for 10-15 cycles with [32P]-dCTP and primers for renin angiotensinogen, after which EF-1 alpha primers were added. Amplified DNA was separated by electrophoresis on polyacrylamide gel and radioactivity in the bands was quantified by direct radioanalytical scanning. Three conditions were necessary to obtain absolute quantification of renin and angiotensinogen mRNA levels: (a) exogenous competitor RNA was used to control for tube-to-tube variability in the efficiencies of reverse transcription and amplification; (b) Sample RNA was titrated with flanking concentrations of competitor RNA to correct for intraassay differences in the efficiency of amplification due to concentration differences between competitor and target templates; and (c) a housekeeping transcript EF-1 alpha was used to control for tube-to-tube differences in RNA loading and/or degradation. We show that the multiplex RT-PCR method is precise and accurate over approximately three logs of transcript concentration and sensitive to less than 5 and 0.5 fg for renin and angiotensinogen mRNA, respectively. This method will be useful for absolute quantification of target mRNAs, especially when the amount of sample RNA is limited or unknown and/or the gene expression is low.


Assuntos
Angiotensinogênio/genética , Reação em Cadeia da Polimerase/métodos , RNA Mensageiro/análise , RNA Mensageiro/genética , Renina/genética , Animais , Sequência de Bases , Técnicas de Cultura , Primers do DNA/genética , Cinética , Masculino , Dados de Sequência Molecular , Fator 1 de Elongação de Peptídeos , Fatores de Alongamento de Peptídeos/genética , Reação em Cadeia da Polimerase/estatística & dados numéricos , Ratos , Ratos Sprague-Dawley , Sensibilidade e Especificidade , Deleção de Sequência , Distribuição Tecidual
20.
Am J Physiol ; 267(5 Pt 1): C1308-18, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7977694

RESUMO

Angiotensin (ANG) II has been previously shown to stimulate proliferation of neonatal rat cardiac fibroblasts via AT1 receptors. Here we conducted studies to assess involvement in this process of two second messengers linked to AT1 receptors, protein kinase C (PKC) and Ca2+. Several findings argue against a dominant role for PKC in ANG II-induced mitogenesis: 1) [Sar1]ANG II, which produced a modest, transient increase in PKC activity, was equally effective in inducing thymidine incorporation into DNA in PKC-depleted cells, whereas the effect of platelet-derived growth factor (PDGF)-BB on thymidine incorporation was reduced to the level observed with [Sar1]ANG II; 2) phorbol 12-myristate 13-acetate (PMA), a potent PKC stimulator, was ineffective in stimulating thymidine incorporation; and 3) PKC downregulation or the highly specific PKC inhibitor, compound 3, eliminated PMA-induced mitogen-activated protein (MAP) kinase activity but did not affect comparable increases induced by [Sar1]ANG II or PDGF-BB. Increased intracellular Ca2+ may be sufficient to account for [Sar1]ANG II-induced MAP kinase activity because ionomycin also increased MAP kinase activity and chelation of intracellular Ca2+ eliminated [Sar1]ANG II-induced activity in PKC-depleted fibroblasts. However, Ca2+ chelation did not prevent [Sar1]ANG II-induced MAP kinase activity in non-PKC-depleted fibroblasts. Thus ANG II can activate MAP kinase in cardiac fibroblasts by either Ca(2+)- or PKC-dependent pathways, and whereas the full effect of PDGF-BB on thymidine incorporation and cell proliferation requires a phorbol ester-sensitive PKC, the hyperplastic growth effect of ANG II does not.


Assuntos
Angiotensina II/farmacologia , Cálcio/fisiologia , Mitose , Miocárdio/citologia , Proteína Quinase C/fisiologia , Animais , Becaplermina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Células Cultivadas , DNA/biossíntese , Ativação Enzimática , Fibroblastos/citologia , Isoenzimas/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-sis , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes , Acetato de Tetradecanoilforbol/farmacologia , Timidina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...