Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Toxicol Sci ; 153(1): 39-54, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27255383

RESUMO

Histone deacetylase (HDAC) inhibitors are an emerging class of anticancer agents that modify gene expression by altering the acetylation status of lysine residues of histone proteins, thereby inducing transcription, cell cycle arrest, differentiation, and cell death or apoptosis of cancer cells. In the clinical setting, treatment with HDAC inhibitors has been associated with delayed cardiac repolarization and in rare instances a lethal ventricular tachyarrhythmia known as torsades de pointes. The mechanism(s) of HDAC inhibitor-induced effects on cardiac repolarization is unknown. We demonstrate that administration of structurally diverse HDAC inhibitors to dogs causes delayed but persistent increases in the heart rate corrected QT interval (QTc), an in vivo measure of cardiac repolarization, at timepoints far removed from the Tmax for parent drug and metabolites. Transcriptional profiling of ventricular myocardium from dogs treated with various HDAC inhibitors demonstrated effects on genes involved in protein trafficking, scaffolding and insertion of various ion channels into the cell membrane as well as genes for specific ion channel subunits involved in cardiac repolarization. Extensive in vitro ion channel profiling of various structural classes of HDAC inhibitors (and their major metabolites) by binding and acute patch clamp assays failed to show any consistent correlations with direct ion channel blockade. Drug-induced rescue of an intracellular trafficking-deficient mutant potassium ion channel, hERG (G601S), and decreased maturation (glycosylation) of wild-type hERG expressed by CHO cells in vitro correlated with prolongation of QTc intervals observed in vivo The results suggest that HDAC inhibitor-induced prolongation of cardiac repolarization may be mediated in part by transcriptional changes of genes required for ion channel trafficking and localization to the sarcolemma. These data have broad implications for the development of these drug classes and suggest that the optimal time to assess potentially transcriptionally mediated physiologic effects will be delayed relative to an epigenetic drug's Tmax/Cmax.


Assuntos
Coração/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Transcrição Gênica , Animais , Transporte Biológico , Cães , Coração/fisiologia , Inibidores de Histona Desacetilases/farmacocinética , Masculino
2.
Artigo em Inglês | MEDLINE | ID: mdl-26427954

RESUMO

INTRODUCTION: Cardiac contractility was evaluated using standard inotropic agents in rats. We compared indices of cardiac contractility, i.e. LV dP/dt max from telemetry while simultaneously collecting EF (ejection fraction) and FS (fractional shortening) measures from echocardiography. METHODS: Male Wistar rats were instrumented with telemetry devices for measurements of blood pressure and left ventricular pressure. Milrinone (PDE III inhibitor) and verapamil (L-type calcium channel blocker) at doses of 0, 3, 10, and 30 mg/kg were administered orally using a 4 × 4 Latin square crossover study design. Telemetry data were recorded at predose and continuously for 24h post-dose. Echocardiographic evaluations were conducted once at predose and at 1 and 2h after milrinone or verapamil administration, respectively. During the recording of echocardiograms, telemetry data were collected simultaneously. Blood samples were also collected to confirm plasma drug exposure. RESULTS: As expected, milrinone increased LV dP/dt max, EF and FS while verapamil decreased LV dP/dt max, EF and FS. Linear regression analysis showed a positive correlation between LV dP/dt max and EF or FS (P<0.001) with both test agents. A change in LV dP/dt max of 1000 mmHg/s was found to correspond with a change in EF and FS of 13 and 16%, respectively, in the telemetered rat. DISCUSSION: The correlation between contractility indices assessed by telemetry and echocardiographic methods in rat models has not received much attention to date. Our results with two reference compounds demonstrate that both methods are sensitive to alterations in contractility induced by inotropic agents administered to rats. The high degree of correlation between changes in LV dP/dt max and EF or FS in the rat enables a translational-element of clinical relevance following changes in contractility indices when measured with telemetry devices in preclinical studies.


Assuntos
Contração Miocárdica/fisiologia , Função Ventricular Esquerda/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Cardiotônicos/farmacologia , Fármacos Cardiovasculares/farmacologia , Estudos Cross-Over , Ecocardiografia/métodos , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Masculino , Milrinona/farmacologia , Contração Miocárdica/efeitos dos fármacos , Ratos , Ratos Wistar , Telemetria/métodos , Função Ventricular Esquerda/efeitos dos fármacos , Pressão Ventricular/efeitos dos fármacos , Pressão Ventricular/fisiologia , Verapamil/farmacologia
3.
PLoS One ; 10(6): e0127498, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26098886

RESUMO

Englerin A is a structurally unique natural product reported to selectively inhibit growth of renal cell carcinoma cell lines. A large scale phenotypic cell profiling experiment (CLiP) of englerin A on ¬over 500 well characterized cancer cell lines showed that englerin A inhibits growth of a subset of tumor cell lines from many lineages, not just renal cell carcinomas. Expression of the TRPC4 cation channel was the cell line feature that best correlated with sensitivity to englerin A, suggesting the hypothesis that TRPC4 is the efficacy target for englerin A. Genetic experiments demonstrate that TRPC4 expression is both necessary and sufficient for englerin A induced growth inhibition. Englerin A induces calcium influx and membrane depolarization in cells expressing high levels of TRPC4 or its close ortholog TRPC5. Electrophysiology experiments confirmed that englerin A is a TRPC4 agonist. Both the englerin A induced current and the englerin A induced growth inhibition can be blocked by the TRPC4/C5 inhibitor ML204. These experiments confirm that activation of TRPC4/C5 channels inhibits tumor cell line proliferation and confirms the TRPC4 target hypothesis generated by the cell line profiling. In selectivity assays englerin A weakly inhibits TRPA1, TRPV3/V4, and TRPM8 which suggests that englerin A may bind a common feature of TRP ion channels. In vivo experiments show that englerin A is lethal in rodents near doses needed to activate the TRPC4 channel. This toxicity suggests that englerin A itself is probably unsuitable for further drug development. However, since englerin A can be synthesized in the laboratory, it may be a useful chemical starting point to identify novel modulators of other TRP family channels.


Assuntos
Proliferação de Células/efeitos dos fármacos , Sesquiterpenos de Guaiano/farmacologia , Canais de Cátion TRPC/agonistas , Animais , Antineoplásicos/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Linhagem Celular Tumoral , Células HEK293 , Humanos , Indóis/farmacologia , Neoplasias Renais/tratamento farmacológico , Camundongos , Camundongos Nus , Piperidinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Ratos , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/genética , Transfecção
4.
Toxicol Sci ; 142(2): 427-35, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25246669

RESUMO

It is widely accepted that more needs to be done to bring new, safe, and efficacious drugs to the market. Cardiovascular toxicity detected both in early drug discovery as well as in the clinic, is a major contributor to the high failure rate of new molecules. The growth of translational safety offers a promising approach to improve the probability of success for new molecules. Here we describe a cross-company initiative to determine the concordance between the conscious telemetered dog and phase I outcome for 3 cardiovascular parameters. The data indicate that, in the context of the methods applied in this analysis, the ability to detect compounds that affect the corrected QT interval (QTc) was good within the 10-30x exposure range but the predictive or detective value for heart rate and diastolic blood pressure was poor. These findings may highlight opportunities to refine both the animal and the clinical study designs, as well as refocusing the assessment of value of dog cardiovascular assessments beyond phase 1. This investigation has also highlighted key considerations for cross-company data sharing and presents a unique learning opportunity to improve future translational projects.


Assuntos
Ensaios Clínicos Fase I como Assunto/estatística & dados numéricos , Avaliação Pré-Clínica de Medicamentos/estatística & dados numéricos , Indústria Farmacêutica/normas , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Pesquisa Translacional Biomédica/métodos , Animais , Pressão Sanguínea/efeitos dos fármacos , Cardiotoxicidade , Ensaios Clínicos Fase I como Assunto/métodos , Ensaios Clínicos Fase I como Assunto/normas , Cães , Avaliação Pré-Clínica de Medicamentos/métodos , Avaliação Pré-Clínica de Medicamentos/normas , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/fisiopatologia , Eletrocardiografia , Voluntários Saudáveis , Frequência Cardíaca/efeitos dos fármacos , Humanos , Valor Preditivo dos Testes , Sensibilidade e Especificidade , Telemetria , Pesquisa Translacional Biomédica/normas , Pesquisa Translacional Biomédica/estatística & dados numéricos
5.
Heart Rhythm ; 9(10): 1706-15, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22710481

RESUMO

BACKGROUND: The ventricular components (QRS and QT) on the electrocardiogram (ECG) depend on the properties of ventricular action potentials that can be modulated by drugs via specific ion channels. However, the correlation of ECG ventricular waveforms with underlying ion actions is not well established and has been extensively debated. OBJECTIVE: To conduct a blinded in vitro assessment of the ionic mechanisms for drug-induced ECG changes. METHODS AND RESULTS: Fourteen cardiac and noncardiac drugs with known effects on cardiac ion channels were selected by the study sponsor, and were tested in the rabbit left ventricular wedge preparation with recording of the ECG and contractility. The investigators who performed the experiments and analyzed the data were blinded to names, concentrations, and molecular weights of the drugs. The compounds were prepared by the sponsor and sent to the investigators as 56 stock solutions. The effects of I(Kr), I(Ks), I(Ca,L), I(Na) blocker, and I(KATP) opener on QRS, QT, and T(p-e), were evaluated. Disclosure of the names and concentrations after completion of the study revealed that there were highly correlated ECG changes with underlying ionic mechanisms and proarrhythmic potential of drugs that, respectively, target I(Kr), I(Ks), I(Ca,L), I(Na), and I(KATP). Among ECG parameters, T(p-e) was more useful in differentiating drugs' actions. CONCLUSIONS: Specific electrophysiological action and the consequent proarrhythmic potential of a drug can be accurately determined by analysis of drug-induced changes in ECG in the rabbit left ventricular wedge preparation. Change in T(p-e) provides the most relevant information.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Eletrocardiografia , Contração Miocárdica/efeitos dos fármacos , Animais , Coelhos
6.
Front Pharmacol ; 3: 6, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22303294

RESUMO

The human cardiac sodium channel (hNav1.5, encoded by the SCN5A gene) is critical for action potential generation and propagation in the heart. Drug-induced sodium channel inhibition decreases the rate of cardiomyocyte depolarization and consequently conduction velocity and can have serious implications for cardiac safety. Genetic mutations in hNav1.5 have also been linked to a number of cardiac diseases. Therefore, off-target hNav1.5 inhibition may be considered a risk marker for a drug candidate. Given the potential safety implications for patients and the costs of late stage drug development, detection, and mitigation of hNav1.5 liabilities early in drug discovery and development becomes important. In this review, we describe a pre-clinical strategy to identify hNav1.5 liabilities that incorporates in vitro, in vivo, and in silico techniques and the application of this information in the integrated risk assessment at different stages of drug discovery and development.

7.
J Pharmacol Exp Ther ; 326(2): 443-52, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18499744

RESUMO

The transient receptor potential (TRP) vanilloid subtype 4 (V4) is a nonselective cation channel that exhibits polymodal activation and is expressed in the endothelium, where it contributes to intracellular Ca2+ homeostasis and regulation of cell volume. The purpose of the present study was to evaluate the systemic cardiovascular effects of GSK1016790A, a novel TRPV4 activator, and to examine its mechanism of action. In three species (mouse, rat, and dog), the i.v. administration of GSK1016790A induced a dose-dependent reduction in blood pressure, followed by profound circulatory collapse. In contrast, GSK1016790A had no acute cardiovascular effects in the TRPV4-/- null mouse. Hemodynamic analyses in the dog and rat demonstrate a profound reduction in cardiac output. However, GSK1016790A had no effect on rate or contractility in the isolated, buffer-perfused rat heart, and it produced potent endothelial-dependent relaxation of rodent-isolated vascular ring segments that were abolished by nitric-oxide synthase (NOS) inhibition (N-nitro-L-arginine methyl ester; L-NAME), ruthenium red, and endothelial NOS (eNOS) gene deletion. However, the in vivo circulatory collapse was not altered by NOS inhibition (L-NAME) or eNOS gene deletion but was associated with (concentration and time appropriate) profound vascular leakage and tissue hemorrhage in the lung, intestine, and kidney. TRPV4 immunoreactivity was localized in the endothelium and epithelium in the affected organs. GSK1016790A potently induced rapid electrophysiological and morphological changes (retraction/condensation) in cultured endothelial cells. In summary, inappropriate activation of TRPV4 produces acute circulatory collapse associated with endothelial activation/injury and failure of the pulmonary microvascular permeability barrier. It will be important to determine the role of TRPV4 in disorders associated with edema and microvascular congestion.


Assuntos
Aorta Torácica/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Leucina/análogos & derivados , Sulfonamidas/efeitos adversos , Canais de Cátion TRPV/agonistas , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Aorta Torácica/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Cães , Relação Dose-Resposta a Droga , Endotélio Vascular/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Leucina/efeitos adversos , Leucina/farmacocinética , Masculino , Camundongos , Camundongos Knockout , Estrutura Molecular , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Sulfonamidas/farmacocinética , Canais de Cátion TRPV/genética , Vasoconstrição/efeitos dos fármacos
8.
Mol Cell Biochem ; 295(1-2): 113-20, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16896535

RESUMO

Lysophosphatidylcholine (LPC) is the major bioactive lipid component of oxidized LDL, thought to be responsible for many of the inflammatory effects of oxidized LDL described in both inflammatory and endothelial cells. Inflammation-induced transformation of vascular smooth muscle cells from a contractile phenotype to a proliferative/secretory phenotype is a hallmark of the vascular remodeling that is characteristic of atherogenesis; however, the role of LPC in this process has not been fully described. The present study tested the hypothesis that LPC is an inflammatory stimulus in coronary artery smooth muscle cells (CASMCs). In cultured human CASMCs, LPC stimulated time- and concentration-dependent release of arachidonic acid that was sensitive to phospholipase A2 and C inhibition. LPC stimulated the release of arachidonic acid metabolites leukotriene-B4 and 6-keto-prostaglandin F1alpha, within the same time course. LPC was also found to stimulate basic fibroblast growth factor release as well as stimulating the release of the cytokines GM-CSF, IL-6, and IL-8. Optimal stimulation of these signals was obtained via palmitic acid-substituted LPC species. Stimulation of arachidonic acid, inflammatory cytokines and growth factor release, implies that LPC might play a multifactorial role in the progression of atherosclerosis, by affecting inflammatory processes.


Assuntos
Vasos Coronários/citologia , Vasos Coronários/efeitos dos fármacos , Inflamação/patologia , Lisofosfatidilcolinas/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , 6-Cetoprostaglandina F1 alfa/biossíntese , Ácido Araquidônico/metabolismo , Ácidos Araquidônicos/farmacologia , Células Cultivadas , Citocinas/metabolismo , Fator 2 de Crescimento de Fibroblastos/biossíntese , Humanos , Leucotrieno B4/biossíntese , Lipídeos/farmacologia , Miócitos de Músculo Liso/metabolismo , Fosfolipases/antagonistas & inibidores , Trítio
9.
J Pharmacol Toxicol Methods ; 54(2): 106-15, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16650782

RESUMO

INTRODUCTION: Methods most commonly used for detecting susceptibility to orthostatic hypotension in humans include head-up tilt and the application of lower body negative pressure (LBNP). The objective of this study was to evaluate the use of LBNP for detecting drug-induced changes in susceptibility to orthostatic hypotension in conscious monkeys (Macaca fascicularis). METHODS: Orthostatic responses were produced using an airtight chamber, which sealed around the stomach (umbilical area) and enclosed the lower body, to which were applied successive decrements of 10 mmHg chamber pressure every 5 min until the orthostatic response was observed. Cardiovascular measurements, involving arterial pressures, heart rate, and left ventricular pressures were recorded. The hypotensive agents prazosin and minoxidil were administered to evaluate the ability of the procedure to detect drug-induced changes in the susceptibility to orthostatic hypotension. RESULTS: A rapid decrease in systolic arterial pressure of >20 mmHg occurring within a 30 s time period was determined to be the best indicator of an orthostatic response. The application of LBNP produced an orthostatic response in all monkeys and on all occasions (100% response). The onset, rate and magnitude of the decrease in systolic blood pressure were also consistent for each monkey. Prazosin (>or=0.16 mg/kg, iv) produced an increase in the susceptibility to the orthostatic response, whereas minoxidil (10 mg/kg, po) had no effect. These results are consistent with previous findings in humans, where similar decreases in arterial pressures occur following the administration of prazosin and minoxidil, whereas increased susceptibility to orthostatic hypotension only occurs with prazosin. DISCUSSION: The results of this study demonstrate that the application of the LBNP is a reliable method for producing an orthostatic hypotensive response in conscious monkeys. In addition, the use of positive (prazosin) and negative (minoxidil) controls demonstrated that the use of LBNP is a valid method for evaluating the effect of drug treatment on susceptibility to orthostatic hypotension.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Hipotensão Ortostática/induzido quimicamente , Hipotensão Ortostática/fisiopatologia , Pressão Negativa da Região Corporal Inferior , Animais , Anti-Hipertensivos/toxicidade , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Frequência Cardíaca/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Macaca fascicularis , Masculino , Minoxidil/toxicidade , Prazosina/toxicidade , Reprodutibilidade dos Testes
10.
J Pharmacol Exp Ther ; 307(3): 932-8, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14561851

RESUMO

Numerous mediators, believed to play a role in endothelial dysfunction (e.g., neurohormones, cytokines, hypoxia, and stretch), have been shown to activate p38 mitogen-activated protein kinase (MAPK) in a variety of cell types. The purpose of the present study was to examine the regulation of p38 MAPK in endothelium and its role in endothelial dysfunction and salt sensitivity. In cultured human umbilical vein endothelial cells (HUVECs), tumor necrosis factor-alpha and lipopolysaccharide increased phosphorylation of p38 MAPK (P-p38 MAPK) and increased ICAM-1 expression. Preincubation with highly selective p38 MAPK inhibitors, 1-(1,3-dihydroxyprop-2-yl)-4-(4-fluorophenyl)-5-[2-phenoxypyrimidin-4-yl] imidazole (SB-239063AN) or SB-239063, dose dependently reduced intercellular adhesion molecule-1 expression in HUVECs. In spontaneously hypertensive-stroke prone rats (SHR-SP), P-p38 MAPK was localized by immunohistochemistry to the aortic endothelium and adventitia but was undetectable in aortae from normotensive rats. Introduction of a salt/fat diet (SFD) to the SHR-SP strain induced endothelial dysfunction (ex vivo vascular reactivity analysis), albuminuria, and an increase in blood pressure within 4 weeks. Chronic dietary dosing (approx. 100 mg/kg/day) with SB-239063AN inhibited the SFD diet-induced hypertension. In addition, delayed treatment also significantly improved survival and restored nitric oxide-mediated endothelium-dependent relaxation in SFD-SHR-SPs with established endothelial dysfunction. These results suggest an important role for p38 MAPK in endothelial inflammation and dysfunction as well as providing the first evidence for p38 MAPK-dependent hypertension.


Assuntos
Endotélio Vascular/patologia , Inibidores Enzimáticos/uso terapêutico , Hipertensão/tratamento farmacológico , Hipertensão/patologia , Imidazóis/farmacologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Pirimidinas/farmacologia , Albuminúria/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Western Blotting , Carbacol/farmacologia , Moléculas de Adesão Celular/biossíntese , Humanos , Imuno-Histoquímica , Masculino , Agonistas Muscarínicos/farmacologia , Ratos , Ratos Endogâmicos SHR , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia , Sobrevida , Telemetria , Proteínas Quinases p38 Ativadas por Mitógeno
11.
Curr Opin Investig Drugs ; 4(9): 1059-64, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14582449

RESUMO

p38 Mitogen-activated protein kinase (MAPK) has been implicated in cardiovascular disease and is activated by various factors, including neurohormones (e.g., catecholamines, angiotensin II and endothelin), hypoxia and wall stress. Activation of p38 MAPK can cause cardiac hypertrophy, negative inotropy and endothelial dysfunction. All of these conditions lead to chronic cardiovascular disease, which is becoming an ever growing burden on society. p38 MAPK inhibition may therefore be an interesting therapeutic approach to the treatment of various cardiovascular diseases. However, in vitro and in vivo results are conflicting and caution must be applied in the translation of bench results to the clinic.


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Animais , Doenças Cardiovasculares/enzimologia , Doença Crônica , Ensaios Clínicos como Assunto , Inibidores Enzimáticos/química , Coração/efeitos dos fármacos , Humanos , Estrutura Molecular , Miocárdio/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno
12.
J Pharmacol Exp Ther ; 301(1): 15-20, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11907152

RESUMO

The vascular response to mechanical injury involves inflammatory and fibroproliferative processes that result in the formation of neointima and vascular remodeling. The complex cellular interactions initiated by vascular injury are coordinated and modulated by the elaboration of cytokines and growth factors. The production and transduction of many of these mediators require phosphorylation of p38 mitogen-activated protein kinase (MAPK). In the present investigation, we examined the pattern and localization of p38 MAPK activation following balloon vascular injury. The effects of long-term and selective inhibition of p38 MAPK with SB 239063 (trans-1-(4-hydroxycyclohexyl)-4-(4-fluorophenyl)-5-[2-methoxy)pyrimidin-4-yl]imidazole) were also investigated in a model of vascular injury. Western blotting and immunohistochemical staining demonstrated that phospho-p38 MAPK was increased following balloon injury of the rabbit iliofemoral artery. The p38 MAPK activation was noted as early as 15 min after balloon injury and remained elevated for at least 28 days. Phospho-p38 MAPK immunoreactivity (IR) was localized primarily in regions of dedifferentiated, smooth muscle alpha-actin-positive cells in all lamina of the vessel wall. Phospho-p38 MAPK IR was not correlated with the localization of macrophage or proliferating cells (proliferating cell nuclear antigen; PCNA +). Long-term treatment (4 weeks) with SB 239063 (50 mg/kg/day, p.o.) reduced the vascular response to injury in the hypercholesterolemic rabbit. SB 239063 had no effect on platelet-derived growth factor (PDGF)-stimulated migration or proliferation of rabbit vascular smooth muscle cells (VSMCs) in culture. However, SB 239063 produced a concentration-dependent inhibition of transforming growth factor (TGF)-beta-stimulated fibronectin production in VSMCs. In conclusion, sustained activation of p38 MAPK plays an important role in the vascular response to injury and inhibition of p38 MAPK may represent a novel therapeutic approach to limit this response.


Assuntos
Vasos Sanguíneos/lesões , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neovascularização Patológica/enzimologia , Angioplastia com Balão , Animais , Vasos Sanguíneos/patologia , Western Blotting , Divisão Celular/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Ativação Enzimática/fisiologia , Fibronectinas/biossíntese , Hipercolesterolemia/sangue , Hipercolesterolemia/patologia , Imidazóis/farmacologia , Imuno-Histoquímica , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Neovascularização Patológica/patologia , Pirimidinas/farmacologia , Coelhos , Especificidade por Substrato , Proteínas Quinases p38 Ativadas por Mitógeno
13.
Am J Physiol Heart Circ Physiol ; 282(3): H1071-80, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11834506

RESUMO

We examined the effect of fibroblast growth factor (FGF)-2 on myocardial resistance to injury when administered after the onset of ischemia, in vivo and ex vivo, and the role of FGF-2 receptors and protein kinase C (PKC). FGF-2 was injected into the left ventricle of rats undergoing permanent surgical coronary occlusion leading to myocardial infarction (MI). After 24 h, FGF-2-treated hearts displayed significantly reduced injury, determined by histological staining and troponin T release, and improved developed pressure compared with untreated controls. An FGF-2 mutant with diminished affinity for the tyrosine kinase FGF-2 receptor 1 (FGFR1) was not cardioprotective. FGF-2-treated hearts retained improved function and decreased damage at 6 wk after MI. In the ex vivo heart, FGF-2 administration during reperfusion after 30-min ischemia improved functional recovery and increased relative levels of PKC subtypes alpha, epsilon, and zeta in the particulate fraction, in a chelerythrine-preventable mode; it also decreased loss of energy metabolites. We conclude that intramyocardial FGF-2 administration shortly after the onset of ischemia confers protection from acute and chronic cardiac dysfunction and damage; FGF-2 delivered during reperfusion protects from ischemia-reperfusion injury; and protection by FGF-2 requires intact binding to FGFR1 and is likely mediated by PKC.


Assuntos
Fator 2 de Crescimento de Fibroblastos/farmacologia , Infarto do Miocárdio/prevenção & controle , Isquemia Miocárdica/prevenção & controle , Proteína Quinase C/metabolismo , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Animais , Biomarcadores/sangue , Diástole/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/administração & dosagem , Ventrículos do Coração , Injeções , Masculino , Contração Miocárdica/efeitos dos fármacos , Infarto do Miocárdio/fisiopatologia , Isquemia Miocárdica/fisiopatologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Fatores de Tempo , Troponina T/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...