Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Protein Pept Sci ; 7(3): 229-39, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16787262

RESUMO

Corticotropin-releasing factor (CRF), CRF-related peptides and their receptors are present in the central nervous system and in peripheral tissues including the immune, reproductive and cardiovascular systems. CRF and urocortin (urocortin 1) bind to the CRF receptor type 1 (CRF(1) receptor) and the CRF receptor type 2 (CRF(2) receptor), whereas urocortin 2 (formerly known as stresscopin related peptide) and urocortin 3 (formerly known as stresscopin) bind with high affinity to the CRF(2) receptor. Recent studies show that urocortin 1, urocortin 2 and urocortin 3 are potent regulators of cardiovascular function. This review highlights the role of cardiovascular CRF and related peptides and its relevance in mediating the adaptive response of the cardiovascular system to stressful conditions.


Assuntos
Fenômenos Fisiológicos Cardiovasculares , Hormônio Liberador da Corticotropina/fisiologia , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Animais , Sistema Cardiovascular/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Hormônio Liberador da Corticotropina/farmacologia , Glucocorticoides/metabolismo , Humanos , Hormônios Peptídicos/metabolismo , Hormônios Peptídicos/farmacologia , Receptores de Hormônio Liberador da Corticotropina/metabolismo
2.
Circulation ; 104(18): 2216-21, 2001 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-11684634

RESUMO

Background- Left ventricular (LV) remodeling after myocardial infarction (MI) is associated with fibrosis, dilatation, and dysfunction. We postulated that prevention of fibrosis after MI with a prolyl 4-hydroxylase inhibitor (P4HI) would preserve LV function and attenuate LV enlargement. Methods and Results- Adult female rats (200 to 250 g) had experimental MI and were then randomized to treatment with P4HI (MI-FG041, n=29) or vehicle (MI-control, n=29) 48 hours after MI for 4 weeks in 2 phases. Echocardiograms were performed weekly with a 15-MHz linear transducer, and at 4 weeks, collagen isoform determinations and in vivo hemodynamics were performed. At randomization, the infarct size and LV function and size were similar in MI-FG041 and MI-control but significantly different from shams (n=9). At week 4, the LV function in MI-FG041 was significantly better than in MI-controls (fractional shortening 21% versus 16%, P=0.01; fractional area change 30% versus 19%, P=0.002; ejection fraction 35% versus 23%, P=0.001). In the FG041 group, LV area in systole was less (P<0.05), the dP/dt(max) after isoproterenol was higher (P<0.05), and types I and III collagen in noninfarcted LV were less than in MI-control. The hydroxyproline/proline ratio was increased by 64% in MI-control and reduced to the sham value in MI-FG041 rats. In the scar tissue, it was reduced by 24% in MI-FG041. Conclusions- This study demonstrates that prevention of interstitial fibrosis with a P4H inhibitor alters the pattern of LV enlargement and produces partial recovery of LV function after MI.


Assuntos
Colágeno/biossíntese , Dilatação Patológica/prevenção & controle , Inibidores Enzimáticos/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Dilatação Patológica/etiologia , Dilatação Patológica/fisiopatologia , Modelos Animais de Doenças , Ecocardiografia , Inibidores Enzimáticos/sangue , Feminino , Fibrose/patologia , Fibrose/prevenção & controle , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Hemodinâmica/efeitos dos fármacos , Hidroxiprolina/metabolismo , Infarto do Miocárdio/complicações , Infarto do Miocárdio/fisiopatologia , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Prolina/metabolismo , Ratos , Ratos Wistar , Recuperação de Função Fisiológica/efeitos dos fármacos , Volume Sistólico/efeitos dos fármacos
3.
J Mol Cell Cardiol ; 33(10): 1791-803, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11603922

RESUMO

Beta-adrenergic stimulation of ventricular myocytes has been shown to induce apoptosis; however, the cellular mechanisms involved in this pathway have not been completely characterized. This study examines the role of protein kinase C (PKC) in the signaling cascade that mediates beta-adrenergic stimulation-induced apoptosis. Stimulation of beta-adrenergic receptors using isoproterenol (ISO, 1-10 microm, 24 h) induced apoptosis in cultured adult rat ventricular myocytes (ARVM) in a dose-dependent manner. Treatment with ISO significantly resulted in the membrane translocation of PKC(epsilon), but not of PKC alpha or delta in ARVM. The activation of PKC(epsilon) by ISO was confirmed using an immune complex kinase assay. To address whether PKC(epsilon) is involved in the mechanism of ISO-induced apoptosis, we used the PKC(epsilon)-specific translocation inhibitor peptide, epsilonV1-2. Peptide epsilonV1-2 significantly blocked the translocation of PKC(epsilon), as well as the enzymatic action of PKC(epsilon), resulting from ISO stimulation. The inhibition of PKC(epsilon) attenuated ISO-induced apoptosis as measured by terminal deoxynucleotidyltransferase nick-end labeling (TUNEL) assay (18.2+/-3.8%v 49.0+/-2.4%P<0.05), while a PKC delta-specific peptide translocation inhibitor (delta V1-1) failed to do so (39.8+/-7.8%). In the presence of ISO, PKC(epsilon) inhibition by epsilonV1-2 was found to significantly enhance activity of ERK, but not that of Akt/PKB. Inhibition of ERK activation by PD 98059 (10-50 microm) attenuated the epsilonV1-2 peptide-mediated anti-apoptotic effect, thus suggesting that ERK activation is involved in this anti-apoptotic effect. Therefore, our results suggest that activation of PKC(epsilon) downstream of beta-adrenergic stimulation promotes apoptosis largely via inhibition of an ERK activation-dependent anti-apoptotic effect.


Assuntos
Agonistas Adrenérgicos beta/metabolismo , Apoptose , Ventrículos do Coração/enzimologia , Isoenzimas/metabolismo , Isoenzimas/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Quinase C/metabolismo , Proteína Quinase C/fisiologia , Animais , Sobrevivência Celular , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Isoproterenol/farmacologia , Masculino , Peptídeos/farmacologia , Proteína Quinase C-épsilon , Transporte Proteico , Ratos , Ratos Wistar , Transdução de Sinais
4.
Am J Physiol Cell Physiol ; 280(5): C1114-20, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11287324

RESUMO

Activation of protein kinase C (PKC) in heart muscle signals hypertrophy and may also directly affect contractile function. We tested this idea using a transgenic (TG) mouse model in which conditionally expressed PKCbeta was turned on at 10 wk of age and remained on for either 6 or 10 mo. Compared with controls, TG cardiac myocytes demonstrated an increase in the peak amplitude of the Ca(2+) transient, an increase in the extent and rate of shortening, and an increase in the rate of relengthening at both 6 and 10 mo of age. Phospholamban phosphorylation and Ca(2+)-uptake rates of sarcoplasmic reticulum vesicles were the same in TG and control heart preparations. At 10 mo, TG skinned fiber bundles demonstrated the same sensitivity to Ca(2+) as controls, but maximum tension was depressed and there was increased myofilament protein phosphorylation. Our results differ from studies in which PKCbeta was constitutively overexpressed in the heart and in studies that reported a depression of myocyte contraction with no change in the Ca(2+) transient.


Assuntos
Sinalização do Cálcio/fisiologia , Coração/fisiologia , Isoenzimas/metabolismo , Contração Miocárdica/fisiologia , Proteína Quinase C/metabolismo , Envelhecimento , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Regulação Enzimológica da Expressão Gênica , Coração/crescimento & desenvolvimento , Isoenzimas/genética , Camundongos , Camundongos Transgênicos , Miocárdio/citologia , Fosfatos/metabolismo , Fosforilação , Proteína Quinase C/genética , Proteína Quinase C beta
5.
Am J Physiol Heart Circ Physiol ; 280(5): H2264-70, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11299230

RESUMO

Studies in human and rodent models have shown that activation of protein kinase C-beta (PKC-beta) is associated with the development of pathological hypertrophy, suggesting that ablation of the PKC-beta pathway might prevent or reverse cardiac hypertrophy. To explore this, we studied mice with targeted disruption of the PKC-beta gene (knockout, KO). There were no detectable differences in expression or distribution of other PKC isoforms between the KO and control hearts as determined by Western blot analysis. Baseline hemodynamics were measured using a closed-chest preparation and there were no differences in heart rate and arterial or left ventricular pressure. Mice were subjected to two independent hypertrophic stimuli: phenylephrine (Phe) at 20 mg x kg(-1) x day(-1) sq infusion for 3 days, and aortic banding (AoB) for 7 days. KO animals demonstrated an increase in heart weight-to-body weight ratio (Phe, 4.3 +/- 0.6 to 6.1 +/- 0.4; AoB, 4.0 +/- 0.1 to 5.8 +/- 0.7) as well as ventricular upregulation of atrial natriuretic factor mRNA analogous to those seen in control animals. These results demonstrate that PKC-beta expression is not necessary for the development of cardiac hypertrophy nor does its absence attenuate the hypertrophic response.


Assuntos
Cardiomegalia/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Miocárdio/enzimologia , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Animais , Aorta/fisiologia , Fator Natriurético Atrial/genética , Biomarcadores , Pressão Sanguínea/fisiologia , Artérias Carótidas/fisiologia , Regulação Enzimológica da Expressão Gênica , Óperon Lac , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase C beta , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Remodelação Ventricular/fisiologia , beta-Galactosidase
6.
Mol Cell Biochem ; 226(1-2): 83-8, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11768242

RESUMO

Recently, the calcium-calmodulin-dependent calcineurin pathway has been defined as a central pathway for the induction of cardiac hypertrophy. The purpose of this study was to determine if cardiac hypertrophy in animals chronically treated with angiotensin II (AngII), could be prevented by blocking this pathway with cyclosporin A (CsA). Female Wistar rats were treated with AngII by subcutaneous infusion and injected twice a day with CsA (25 mg/kg) for 7 days. In the AngII treated group there was a 30% increase in the heart/body weight ratio (p < 0.05 vs. control). The increase in heart weight was blocked with CsA. Substantial increases in ANF and betaMHC gene expression were detected in the AngII treated animals, which were either attenuated or blocked with CsA treatment. Thus, this study demonstrates that CsA does prevent the development of cardiac hypertrophy in AngII treated rats, suggesting that the calcium-calmodulin-dependent calcineurin pathway is associated with angiotensin II induced hypertrophy in vivo.


Assuntos
Angiotensina II/metabolismo , Cardiomegalia/metabolismo , Ciclosporina/farmacologia , Miocárdio/metabolismo , Animais , Northern Blotting , Peso Corporal , Calcineurina/metabolismo , Cálcio/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Feminino , Hipertrofia , Tamanho do Órgão , RNA/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais , Regulação para Cima
7.
Heart Dis ; 2(6): 409-21, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11728292

RESUMO

It has been slightly more than 10 years since the first proof-of-concept studies were performed, which demonstrated the feasibility of gene transfer into the heart and vasculature of experimental animals. Since that time there has been a dramatic increase in the nature and sophistication of gene transfer techniques and also in the number of cardiovascular diseases that are potential targets for gene-based therapies. In this article, the authors review the current strategies for gene delivery, including viral and nonviral approaches. The authors also highlight several biologic processes within the cardiovascular system, including restenosis, experimental angiogenesis, heart failure, and atherosclerosis-conditions for which gene therapy shows promise. It is hoped that this will provide an update of this therapeutic strategy for the year 2000.


Assuntos
Doenças Cardiovasculares/terapia , Técnicas de Transferência de Genes , Terapia Genética , Doenças Cardiovasculares/genética , Humanos , Técnicas In Vitro
8.
Proc Natl Acad Sci U S A ; 96(23): 13536-41, 1999 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-10557356

RESUMO

Activation of protein kinase C (PKC) protects the heart from ischemic injury; however, its mechanism of action is unknown, in part because no model for chronic activation of PKC has been available. To test whether chronic, mild elevation of PKC activity in adult mouse hearts results in myocardial protection during ischemia or reperfusion, hearts isolated from transgenic mice expressing a low level of activated PKCbeta throughout adulthood (beta-Tx) were compared with control hearts before ischemia, during 12 or 28 min of no-flow ischemia, and during reperfusion. Left-ventricular-developed pressure in isolated isovolumic hearts, normalized to heart weight, was similar in the two groups at baseline. However, recovery of contractile function was markedly improved in beta-Tx hearts after either 12 (97 +/- 3% vs. 69 +/- 4%) or 28 min of ischemia (76 +/- 8% vs. 48 +/- 3%). Chelerythrine, a PKC inhibitor, abolished the difference between the two groups, indicating that the beneficial effect was PKC-mediated. (31)P NMR spectroscopy was used to test whether modification of intracellular pH and/or preservation of high-energy phosphate levels during ischemia contributed to the cardioprotection in beta-Tx hearts. No difference in intracellular pH or high-energy phosphate levels was found between the beta-Tx and control hearts at baseline or during ischemia. Thus, long-term modest increase in PKC activity in adult mouse hearts did not alter baseline function but did lead to improved postischemic recovery. Furthermore, our results suggest that mechanisms other than reduced acidification and preservation of high-energy phosphate levels during ischemia contribute to the improved recovery.


Assuntos
Isquemia Miocárdica/fisiopatologia , Miocárdio/enzimologia , Proteína Quinase C/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Concentração de Íons de Hidrogênio , Espectroscopia de Ressonância Magnética , Camundongos , Camundongos Transgênicos , Isquemia Miocárdica/enzimologia , Isquemia Miocárdica/metabolismo , Miocárdio/metabolismo , Fosfocreatina/metabolismo
9.
J Biol Chem ; 274(21): 15030-40, 1999 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-10329706

RESUMO

The paradigm for the response to hypoxia is erythropoietin gene expression; activation of hypoxia-inducible factor-1 (HIF-1) results in erythropoietin production. Previously, we found that oxygen deprivation induced tissue factor, especially in mononuclear phagocytes, by an early growth response (Egr-1)-dependent pathway without involvement of HIF-1 (Yan, S.-F., Zou, Y.-S., Gao, Y., Zhai, C., Mackman, N., Lee, S., Milbrandt, J., Pinsky, D., Kisiel, W., and Stern, D. (1998) Proc. Natl. Acad. Sci. U. S. A. 95, 8298-8303). Now, we show that cultured monocytes subjected to hypoxia (pO2 approximately 12 torr) displayed increased Egr-1 expression because of de novo biosynthesis, with a approximately 10-fold increased rate of transcription. Transfection of monocytes with Egr-1 promoter-luciferase constructs localized elements responsible for hypoxia-enhanced expression to -424/-65, a region including EBS (ets binding site)-SRE (serum response element)-EBS and SRE-EBS-SRE sites. Further studies with each of these regions ligated to the basal thymidine kinase promoter and luciferase demonstrated that EBS sites in the element spanning -424/-375 were critical for hypoxia-enhanceable gene expression. These data suggested that an activated ets factor, such as Elk-1, in complex with serum response factor, was the likely proximal trigger of Egr-1 transcription. Indeed, hypoxia induced activation of Elk-1, and suppression of Elk-1 blocked up-regulation of Egr-1 transcription. The signaling cascade preceding Elk-1 activation in response to oxygen deprivation was traced to activation of protein kinase C-betaII, Raf, mitogen-activated protein kinase/extracellular signal-regulated protein kinase kinase and mitogen-activated protein kinases. Comparable hypoxia-mediated Egr-1 induction and activation were observed in cultured hepatoma-derived cells deficient in HIF-1beta and wild-type hepatoma cells, indicating that the HIF-1 and Egr-1 pathways are initiated independently in response to oxygen deprivation. We propose that activation of Egr-1 in response to hypoxia induces a different facet of the adaptive response than HIF-1, one component of which causes expression of tissue factor, resulting in fibrin deposition.


Assuntos
Hipóxia Celular/genética , Proteínas de Ligação a DNA/fisiologia , Proteínas Imediatamente Precoces/fisiologia , MAP Quinase Quinase Quinase 1 , Fatores de Transcrição/fisiologia , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Células Cultivadas , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Proteína 1 de Resposta de Crescimento Precoce , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Proteínas Imediatamente Precoces/biossíntese , Proteínas Imediatamente Precoces/genética , Isoenzimas/fisiologia , Proteínas Nucleares/fisiologia , Proteína Quinase C/fisiologia , Proteína Quinase C beta , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-raf/fisiologia , RNA Mensageiro/biossíntese , Ratos , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
10.
Am J Physiol ; 275(3): H961-8, 1998 09.
Artigo em Inglês | MEDLINE | ID: mdl-9724301

RESUMO

To establish whether catecholamines per se in the absence of significant increases in systolic load induce myocardial damage via apoptosis, rats were treated with vehicle or isoproterenol (400 microg . kg-1 . h-1). Apoptotic cardiocytes (Apo) were identified in paraffin-embedded sections using terminal deoxynucleotide transferase-mediated dUTP nick end labeling. Results were confirmed using an independent ligase assay. Systolic blood pressures were comparable in isoproterenol-treated and control rats. Twenty-four hours of treatment with isoproterenol resulted in significant numbers of Apo compared with control [7.9 +/- 2.5 vs. 0.3 +/- 0.3 (SE) cm-2, P < 0.05]. A cohort of animals was subjected to ventricular pacing to induce a tachycardia equivalent to that induced by isoproterenol, and these animals did not show an increase in Apo. The left ventricular hypertrophy induced by 2 wk of abdominal aortic banding also increased Apo ( approximately 7. 2-fold); however, 24 h of isoproterenol infusion did not induce additional Apo in these rats. Thus catecholamines, in the absence of altered systolic load, induce Apo which is not mediated solely by tachycardia. Left ventricular hypertrophy secondary to abdominal aortic banding is associated with Apo, but this does not increase sensitivity to isoproterenol-induced Apo.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Apoptose , Hipertrofia Ventricular Esquerda/patologia , Miocárdio/patologia , Receptores Adrenérgicos beta/fisiologia , Taquicardia/patologia , Animais , Aorta Abdominal , Estimulação Cardíaca Artificial , Constrição , DNA Ligases/metabolismo , DNA Nucleotidilexotransferase/metabolismo , Nucleotídeos de Desoxiuracil , Frequência Cardíaca , Isoproterenol/farmacologia , Masculino , Ratos , Ratos Wistar , Receptores Adrenérgicos beta/efeitos dos fármacos
11.
Circ Res ; 81(6): 1027-33, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9400384

RESUMO

A cardiomyopathy that is characterized by an impairment in diastolic relaxation and a loss of calcium sensitivity of the isolated myofibril has been described in chronic diabetic animals and humans. To explore a possible role for protein kinase C (PKC)-mediated phosphorylation of myofibrillar proteins in this process, we characterized the subcellular distribution of the major PKC isoforms seen in the adult heart in cardiocytes isolated from diabetic rats and determined patterns of phosphorylation of the major regulatory proteins, including troponin I (TnI). Rats were made diabetic with a single injection of streptozotocin, and myocardiocytes were isolated and studied 3 to 4 weeks later. In nondiabetic animals, 76% of the PKC epsilon isoform was located in the cytosol and 24% was particulate, whereas in diabetic animals, 55% was cytosolic and 45% was particulate (P < .05). PKC delta, the other major PKC isoform seen in adult cardiocytes, did not show a change in subcellular localization. In parallel, TnI phosphorylation was increased 5-fold in cardiocytes isolated from the hearts of diabetic animals relative to control animals (P < .01). The change in PKC epsilon distribution and in TnI phosphorylation in diabetic animals was completely prevented by rendering the animals euglycemic with insulin or by concomitant treatment with a specific angiotensin II type-1 receptor (AT1) antagonist. Since PKC phosphorylation of TnI has been associated with a loss of calcium sensitivity of intact myofibrils, these data suggest that angiotensin II receptor-mediated activation of PKC may play a role in the contractile dysfunction seen in chronic diabetes.


Assuntos
Antagonistas de Receptores de Angiotensina , Diabetes Mellitus Experimental/metabolismo , Isoenzimas/metabolismo , Miocárdio/metabolismo , Proteína Quinase C/metabolismo , Troponina I/metabolismo , Animais , Feminino , Imidazóis/farmacologia , Fosforilação , Ratos , Ratos Wistar , Estreptozocina , Tetrazóis/farmacologia
12.
J Clin Invest ; 100(9): 2189-95, 1997 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9410895

RESUMO

Protein kinase C (PKC) activation in the heart has been linked to a hypertrophic phenotype and to processes that influence contractile function. To establish whether PKC activation is sufficient to induce an abnormal phenotype, PKCbeta was conditionally expressed in cardiomyocytes of transgenic mice. Transgene expression in adults caused mild and progressive ventricular hypertrophy associated with impaired diastolic relaxation, whereas expression in newborns caused sudden death associated with marked abnormalities in the regulation of intracellular calcium. Thus, the PKC signaling pathway in cardiocytes has different effects depending on the timing of expression and, in the adult, is sufficient to induce pathologic hypertrophy.


Assuntos
Cardiomegalia/enzimologia , Isoenzimas/metabolismo , Miocárdio/enzimologia , Proteína Quinase C/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Peso Corporal , Cálcio/fisiologia , Morte Súbita , Feminino , Isoproterenol/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Quinase C beta , Sarcômeros/fisiologia
13.
J Mol Cell Cardiol ; 29(10): 2711-6, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9344765

RESUMO

To assess the role of intermittent beta-adrenergic stimulation on alpha-myosin heavy chain expression and cellular hypertrophy, we studied the effect of intermittent dobutamine on myosin heavy chain isoform distribution and protein synthesis in the heterotopic rat heart preparation. This model allows the analysis of a pharmocologic stimulus in isolation from the mechanical load on the myocardium induced by the drug. Intermittent administration of dobutamine resulted in elevated alpha-MHC levels (75 +/- 12%) compared to control (55 +/- 10%; X +/- s.e.; P<0.05) transplanted hearts. This effect was not altered by alpha-receptor blockade with terazosin (72 +/- 8%). Intermittently pacing the transplanted hearts at the same rate as observed with dobutamine alone, also elevated alpha-MHC levels (70 +/- 5%). In contrast, total protein synthesis in the transplanted hearts was not altered with any of the drug or pacing interventions compared to control hearts. These data suggest that intermittent beta-receptor stimulation and/or intermittent increased heart rate contribute to altered patterns of myosin heavy chain expression. However, increases in cardiac mass and protein synthesis are probably mediated by hemodynamic factors rather than catecholamine stimulation.


Assuntos
Catecolaminas/farmacologia , Coração/efeitos dos fármacos , Miocárdio/metabolismo , Cadeias Pesadas de Miosina/biossíntese , Miosinas/biossíntese , Agonistas alfa-Adrenérgicos/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Dobutamina/farmacologia , Feminino , Coração/fisiologia , Frequência Cardíaca/efeitos dos fármacos , Transplante de Coração , Prazosina/análogos & derivados , Prazosina/farmacologia , Biossíntese de Proteínas , Ratos , Ratos Endogâmicos F344
14.
Am J Physiol ; 272(4 Pt 2): H1636-42, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9139945

RESUMO

We recently identified a developmental decline in protein kinase C (PKC) isoform expression, at the level of the protein, in rat ventricular myocardium. To investigate mechanisms regulating PKC isoform expression in cardiac tissue, this study uses Northern blot analysis to compare the abundance of PKC isoform mRNAs in neonatal and adult rat ventricular myocardium. PKC-epsilon protein and mRNA were detected in both neonatal and adult rat ventricular myocardial preparations. In contrast, coordinate postnatal declines in the abundance of PKC-alpha and PKC-delta proteins and transcripts were identified. An antiserum raised against the COOH-terminal sequence of PKC-zeta detected abundant immunoreactivity in neonatal, but not adult, ventricular myocytes. However, PKC-zeta transcripts were not detectable in the heart either by Northern blot analysis or a reverse transcriptase-polymerase chain reaction approach, indicating that neither the myocytes nor the contaminating cellular elements in the heart express PKC-zeta. Rather, PKC-lambda, another atypical PKC isoform that is structurally highly homologous to PKC-zeta, was detected at the protein and mRNA level in neonatal, but not adult, ventricular myocardium. Taken together, these results establish that developmental declines in calcium-sensitive, novel, and atypical PKC isoforms are paralleled by changes in the levels of the mRNAs encoding these proteins, suggesting transcriptional regulation of PKC during normal cardiac development. The results of this study further identify PKC-lambda as the atypical PKC isoform expressed by the immature ventricle.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Isoenzimas/biossíntese , Miocárdio/enzimologia , Proteína Quinase C/biossíntese , Animais , Animais Recém-Nascidos , Feto , Regulação Enzimológica da Expressão Gênica , Coração/crescimento & desenvolvimento , Ventrículos do Coração , Reação em Cadeia da Polimerase , Proteína Quinase C-alfa , Proteína Quinase C-delta , Proteína Quinase C-épsilon , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Transcrição Gênica
15.
Am J Physiol ; 272(1 Pt 2): H409-17, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9038963

RESUMO

To evaluate the resistance of physiologically hypertrophied hearts to hypoxic insult, we quantified the development of functional deficits during hypoxia and reoxygenation in hypertrophied hearts from swim-trained female rats and we correlated this with assessment of high-energy phosphate (HEP) metabolites from simultaneous 31P nuclear magnetic resonance (NMR) measurements. Furthermore, in vivo enzymatic studies were carried out with saturation transfer NMR under well-oxygenated perfusion conditions for both beating and KCl-arrested hearts. Finally, in vitro enzymatic assays were performed. During hypoxia, the trained hearts exhibited improved systolic and diastolic function compared with hearts from sedentary animals. After 16 min of hypoxia, left ventricular (LV) developed pressure fell to 9% of baseline in control hearts but to only 21% of baseline in trained hearts (P < 0.01). LV diastolic function was also improved by training, increasing during hypoxia from a baseline of 10 to 71.0 +/- 3.3 mmHg in control hearts and to 55.3 +/- 4.8 mmHg in trained hearts (P < 0.05). Trained hearts also showed more rapid and complete recovery of function during reoxygenation and greater coronary flow per gram of heart throughout the entire protocol. Functional differences were not accompanied by differences in HEP at baseline; moreover, ATP and phosphocreatine (PCr) loss during hypoxia was similar between control and trained hearts, as was the recovery of PCr during reoxygenation. Saturation transfer experiments showed an increase in the forward creatine kinase (CrK) rate constant in trained hearts of 18% while beating, whereas in vitro enzymatic analysis revealed a 16% increase in the ratio of mitochondrial CrK to citrate synthase activity in LV tissue. Thus the relative preservation of function in hearts from trained rats could not be accounted for by overall HEP levels but may reflect adaptations in the CrK system.


Assuntos
Metabolismo Energético , Coração/fisiologia , Miocárdio/metabolismo , Condicionamento Físico Animal , Trifosfato de Adenosina/metabolismo , Animais , Creatina Quinase/metabolismo , Feminino , Coração/anatomia & histologia , Coração/efeitos dos fármacos , Hipóxia/fisiopatologia , Técnicas In Vitro , Espectroscopia de Ressonância Magnética , Músculo Esquelético/metabolismo , Tamanho do Órgão , Oxigênio/farmacologia , Consumo de Oxigênio , Perfusão , Fosfocreatina/metabolismo , Ratos , Ratos Wistar
16.
J Appl Physiol (1985) ; 81(2): 816-21, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8872651

RESUMO

The renin-angiotensin system has been implicated in the hypertrophic adaptation of the heart to exogenous pathological loads, such as hypertension and aortic stenosis; however, the role of this hormonal system in the cardiac adaptations to physiological loads, such as chronic exercise conditioning, has not been established. We therefore studied the effect of angiotensin receptor 1 (AT1) blockade on the chronic cardiac responses of rats subjected to an 8-wk swimming program. Compared with matched sedentary controls, untreated swimmers increased their left ventricular weights by 13%, and swimmers treated with the AT1 antagonist L-158809 increased their left ventricular weights by 11% (both P < 0.05 vs. sedentary controls). The incorporation of labeled amino acids into the heart at the time of death was unchanged in all groups, and therefore the increase in heart weight in both swim-conditioned groups appeared to reflect a decrease in the rate of protein degradation in the heart. Hearts from both swim-conditioned groups manifested an increase in the V1-predominant myosin isoform pattern but not an increase in atrial natriuretic factor mRNA expression or protein kinase C translocation. The fact that these patterns of adaptation are preserved in exercised conditioned animals treated with an AT1 antagonist suggests that the chronic hypertrophic response of the heart to physiological loads is not influenced by the renin-angiotensin system.


Assuntos
Angiotensina I/metabolismo , Antagonistas de Receptores de Angiotensina , Anti-Hipertensivos/farmacologia , Cardiomegalia/prevenção & controle , Imidazóis/farmacologia , Tetrazóis/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Northern Blotting , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Cardiomegalia/fisiopatologia , Proteínas Contráteis/biossíntese , Feminino , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Tamanho do Órgão/efeitos dos fármacos , Tamanho do Órgão/fisiologia , Condicionamento Físico Animal/fisiologia , Proteína Quinase C/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar
17.
Am J Physiol ; 270(6 Pt 2): H2177-83, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8764271

RESUMO

Myocardial stretch and the renin-angiotensin system have been implicated in the development of cardiac hypertrophy through the activation of specific target genes. However, the relative importance of these putative hypertrophic stimuli has not been established in vivo. We used an isolated isovolumic heart preparation in which coronary perfusion pressure (CPP), left ventricular end-diastolic pressure, and pharmacological therapy can be independently manipulated to study this relationship. High CPP (140 cmH2O), which increased coronary flow (8.99 vs. 17.6 ml/min) and left ventricular systolic pressure (50 vs. 91 mmHg), increased steady state c-fos mRNA expression 2.3-fold (all P < 0.01 vs. low CPP). In contrast, increased left ventricular end-diastolic pressure (25 mmHg) and/or infusion of angiotensin II in the absence of increased CPP was not associated with an increase in c-fos mRNA expression. The change in c-fos gene expression seen with increased CPP was largely reversed by treatment with an angiotensin type 1 (AT1) receptor blocker. Hearts perfused at high CPP demonstrated increased translocation/activation of protein kinase C-epsilon relative to controls. None of the hearts studied were ischemic during perfusion. Thus, in the perfused adult rat heart, dynamic, but not static, stretch activates the early response gene, c-fos, and may involve the endogenous reninangiotensin system and protein kinase C.


Assuntos
Regulação da Expressão Gênica , Isoenzimas/metabolismo , Miocárdio/metabolismo , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Sistema Renina-Angiotensina/fisiologia , Angiotensina II/farmacologia , Antagonistas de Receptores de Angiotensina , Animais , Transporte Biológico/efeitos dos fármacos , Fenômenos Biomecânicos , Feminino , Coração/fisiologia , Técnicas In Vitro , Contração Miocárdica , Proteína Quinase C-épsilon , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar
18.
Am J Physiol ; 267(6 Pt 2): H2149-54, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7810714

RESUMO

The heterotopically transplanted rat heart (TH) undergoes rapid muscle atrophy and a concurrent shift from alpha- to beta-myosin heavy chain (MHC) by 1 wk after surgery. In the current experiments, TH were continuously paced (420 beats/min) for 1 wk beginning 24 h after surgery or for 1 wk beginning 14 days after surgery to determine the role of increased heart rate in preventing or reversing cardiac atrophy. Left ventricular (LV) wet weight (283 vs. 256 mg paced vs. nonpaced) and protein content (32 vs. 23 mg paced vs. nonpaced, P < 0.05) were significantly elevated in TH paced 1 wk after surgery but were unchanged (211 vs. 198 mg and 24 vs. 23 mg LV wet wt and protein content, respectively) in TH paced 2 wk after surgery. Total cardiac protein synthesis in the TH paced immediately after surgery was increased compared with the corresponding nonpaced hearts (5.6 vs. 4.0 mg.mg LV wet wt-1.day-1, P < 0.05), while in the TH, where pacing was initiated 2 wk after surgery, it was unchanged (3.6 vs. 3.7 mg.mg LV wet wt-1.day-1). Fractional synthesis rate was elevated in TH and was not altered by pacing. Pacing the TH also attenuated the shift in alpha-MHC in the first 7 days after surgery but did not reverse the shift 2 wk later. The increase in protein synthesis combined with an unchanged fractional synthesis rate suggests that pacing attenuates cardiac mass by decreasing protein degradation and that once the atrophic process is established, neither synthesis rate nor isomyosin shift can be altered by continuous pacing.


Assuntos
Estimulação Cardíaca Artificial , Transplante de Coração , Miocárdio/metabolismo , Miocárdio/patologia , Miosinas/metabolismo , Animais , Atrofia/prevenção & controle , Feminino , Frequência Cardíaca/fisiologia , Ventrículos do Coração/patologia , Leucina/sangue , Tamanho do Órgão , Biossíntese de Proteínas , RNA de Transferência de Leucina/metabolismo , Ratos , Ratos Endogâmicos F344
19.
Gene Ther ; 1(6): 395-402, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7584105

RESUMO

Important aspects of successful adenovirus gene transfer include the amount and persistence of gene expression, the ability to readminister virus and the localization of virus-directed gene expression to target organs. Our objective in this study was to use a single recombinant adenovirus bearing a quantifiable reporter gene [chloramphenicol acetyltransferase (CAT)] to establish the parameters which define the limits of adenovirus gene expression in a rat model. First, we determined how the route of virus administration affected the amount, duration and distribution of expression in different tissues and in rats of different developmental stages. All routes resulted in infection of all tissues tested. Surprisingly, the most efficient and widespread gene transfer was achieved by intracardiac muscle injection. The high levels of CAT protein that can be produced in a liver (< or = 1.7 mg) or a heart (< or = 196 micrograms) 5 days after infection suggest that the amount of gene product will not be a limitation in the use of adenovirus. Following peak activity at 5 days after infection, a gradual decline of CAT expression was observed in all tissues assayed; by 80 days neither CAT activity nor adenovirus DNA were detectable. In addition, adults could not be boosted by a second administration of virus, presumably due to the presence of high levels of neutralizing antibodies. The limited persistence of gene expression could be circumvented when virus was injected into neonates. Blocking T lymphocyte expansion by cyclosporine enhanced the persistence of CAT gene product over a 25-day period in heart and lung but not in liver compared with control animals.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Animais , Cloranfenicol O-Acetiltransferase/genética , Cloranfenicol O-Acetiltransferase/metabolismo , Expressão Gênica , Genes Reporter , Terapia Genética , Sistema Imunitário/fisiologia , Ratos , Ratos Sprague-Dawley , Recombinação Genética , Linfócitos T/imunologia , Fatores de Tempo , Distribuição Tecidual
20.
Am J Physiol ; 267(3 Pt 2): H1167-73, 1994 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8092282

RESUMO

Transgenic mice can be created to serve as models of human cardiac disease. Despite the technology available to manipulate the cardiovascular system of the mouse, there is relatively little information available concerning the normal physiology of the mouse heart. Therefore, we have characterized the response of the adult mouse to chronic physical conditioning by swimming. Adult female C57/B16 mice were conditioned by swimming up to 90 min twice daily for 4 wk, resulting in a 10% increase in heart weight and a 16% increase in heart weight-to-body weight ratios compared with sedentary controls. The heart rate response to a submaximal work load decreased > 20% with this conditioning program. Succinate dehydrogenase activity increased markedly in the soleus muscles of the conditioned animals, from 28 +/- 3 to 44 +/- 3 nmol.mg-1.min-1. In contrast to these changes, which also characterize the exercise model in the rat, no increase in cardiac tissue norepinephrine content or in cardiac myosin or myofibrillar adenosinetriphosphatase (ATPase) activities was observed, and no change in the V1 predominant myosin isoform or alpha-myosin heavy chain mRNA profiles was seen in the hearts of the swimmers. This study establishes that mice are able to develop cardiac hypertrophy in response to chronic conditioning which is not associated with changes in the ATPase activities of cardiac muscle. These data should be of use to investigators using murine models to define the molecular basis of adaptive cardiac hypertrophy in vivo.


Assuntos
Adaptação Fisiológica , Coração/fisiologia , Condicionamento Físico Animal , Animais , Sequência de Bases , Cardiomegalia/etiologia , Feminino , Frequência Cardíaca , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Músculos/enzimologia , Miosinas/genética , Miosinas/metabolismo , Sondas de Oligonucleotídeos/genética , RNA Mensageiro/metabolismo , Succinato Desidrogenase/metabolismo , Troponina/classificação , Troponina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...