Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Toxicol Rep ; 10: 521-528, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37152411

RESUMO

Dasatinib is a multitargeted kinase inhibitor used for treatment of chronic myeloid leukemia and acute lymphoblastic leukemia. Unfortunately, treatment of cancer patients with some kinase inhibitors has been associated with cardiotoxicity. Cancer treatment with dasatinib has been reported to be associated with cardiotoxic side effects such as left ventricular dysfunction, heart failure, pericardial effusion and pulmonary hypertension. Here we aimed to investigate the molecular mechanisms underlying the cardiotoxicity of dasatinib. We found that among the resident cardiac cell types, cardiomyocytes were most sensitive to dasatinib-induced cell death. Exposure of cardiomyocytes to dasatinib attenuated the activity of extracellular signal-regulated kinase (ERK), which is a downstream target of dasatinib target kinase c-Src. Similar to dasatinib, c-Src depletion in cardiomyocytes compromised cardiomyocyte viability. Overexpression of dasatinib-resistant mutant of c-Src rescued the toxicity of dasatinib on cardiomyocytes, whereas forced expression of wild type c-Src did not have protective effect. Collectively, our results show that c-Src is a key target of dasatinib mediating the toxicity of dasatinib to cardiomyocytes. These findings may influence future drug design and suggest closer monitoring of patients treated with agents targeting c-Src for possible adverse cardiac effects.

2.
Clin Pharmacol Ther ; 114(1): 148-160, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37129347

RESUMO

Since 2006, the European conditional marketing authorization (CMA) aims to facilitate timely patient access to medicinal products for which there is an unmet medical need by accepting less comprehensive data than normally required. The granting of CMA requires a positive benefit-risk balance, unmet medical needs to be fulfilled, likely submission of comprehensive data postauthorization, and the benefit of immediate availability to outweigh the risks of data noncomprehensiveness. Since its first use, more than half of all CMAs represent (hemato-)oncology indications. Therefore, we aimed to investigate the conditions in which CMA has been applied for anticancer medicinal products and whether they have changed over time. We retrospectively assessed the European public assessment reports of the 30 anticancer medicinal products granted CMA in 2006-2020 (51% of all 59 CMAs). Comparison of 2006-2013 to 2014-2020 highlighted increased proportions of proactively requested CMAs (+40%), medicinal products that addressed unmet medical needs by providing a major therapeutic advantage over authorized treatments (+38%), and orphan designated indications (+32%). In contrast, it showed decreased proportions of medicinal products for which a scientific advisory group was consulted (-55%) and phase III randomized controlled trial data were available (-38%). This suggests that applicants and the European Medicines Agency have learned how to use the CMA as a regulatory tool, among others, through better planning and proactive interaction. However, the increasing number of granted CMAs complicates the establishment of unmet medical need and the benefit-risk balance, especially in crowded indications and when only phase II uncontrolled trials are available.


Assuntos
Antineoplásicos , Aprovação de Drogas , Humanos , Estudos Retrospectivos , Antineoplásicos/uso terapêutico , Medição de Risco , Marketing
3.
Clin Pharmacol Ther ; 108(3): 653-660, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32578196

RESUMO

The traditional drug development paradigm, consisting of sequential phases and randomized studies, has been challenged in oncology and hemato-oncology. In the regulatory context, a number of new products have been authorized based on nonrandomized efficacy and safety data. We retrospectively analyzed the European public assessment reports for anticancer treatments authorized between 2010 and 2019 to describe the data behind such approvals. Twenty-two initial marketing authorizations, mainly conditional, were identified. Fifty percent of the products had an orphan indication, and 77% had received previous scientific advice. Conclusions of clinical benefit were based on tumor responses, ranging between 15.8 and 88%. Our data shows that single-arm clinical studies leading to positive regulatory outcomes share common methodological approaches and end points, mostly comparing the overall response rate with a fixed success threshold as the primary analysis. The clinical indications in these approvals are clustered in late-line settings, hematological malignancies, and lung cancer. Our findings underline the need to reflect on the current practice, the methodological aspects, and end points in single-arm studies, and develop specific regulatory guidance on nonrandomized and novel study designs.


Assuntos
Antineoplásicos/uso terapêutico , Ensaios Clínicos como Assunto , Aprovação de Drogas , Projetos de Pesquisa , Antineoplásicos/efeitos adversos , Determinação de Ponto Final , Europa (Continente) , Órgãos Governamentais , Humanos , Segurança do Paciente , Estudos Retrospectivos , Medição de Risco , Fatores de Tempo , Resultado do Tratamento
4.
J Am Heart Assoc ; 8(21): e013018, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31617439

RESUMO

Background Small molecule kinase inhibitors (KIs) are a class of agents currently used for treatment of various cancers. Unfortunately, treatment of cancer patients with some of the KIs is associated with cardiotoxicity, and there is an unmet need for methods to predict their cardiotoxicity. Here, we utilized a novel computational method to identify protein kinases crucial for cardiomyocyte viability. Methods and Results One hundred forty KIs were screened for their toxicity in cultured neonatal cardiomyocytes. The kinase targets of KIs were determined based on integrated data from binding assays. The key kinases mediating the toxicity of KIs to cardiomyocytes were identified by using a novel machine learning method for target deconvolution that combines the information from the toxicity screen and from the kinase profiling assays. The top kinases identified by the model were phosphoinositide 3-kinase catalytic subunit alpha, mammalian target of rapamycin, and insulin-like growth factor 1 receptor. Knockdown of the individual kinases in cardiomyocytes confirmed their role in regulating cardiomyocyte viability. Conclusions Combining the data from analysis of KI toxicity on cardiomyocytes and KI target profiling provides a novel method to predict cardiomyocyte toxicity of KIs.


Assuntos
Sobrevivência Celular , Aprendizado de Máquina , Miócitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptor IGF Tipo 1/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cardiotoxinas/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Miócitos Cardíacos/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Ratos
5.
J Cancer Res Clin Oncol ; 144(8): 1613-1621, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29872916

RESUMO

PURPOSE: Cardiotoxicity is the most important side effect of trastuzumab treatment. Heart function monitoring is recommended during the treatment which has led to growing use of resources. The aim of this retrospective study was to determine the frequency and timing of trastuzumab cardiotoxicity and its risk factors in real-world setting. METHODS: Single institute, retrospective collection of data on HER2+ breast cancer patients (n = 246) was carried out through a pharmacy search for patients who had received trastuzumab in 2006-2014. Clinical and pathological factors, treatment history, EF measurements, cardiac medications, cardiovascular disease history, cardiac symptoms, and survival data were collected from patient records. RESULTS: 32 patients (13%) had EF decline ≥ 10%, eleven (4.5%) had EF decline ≥ 20% within 1 year after trastuzumab initiation, and trastuzumab was discontinued due to suspected cardiotoxicity in six patients (2.4%). 49 patients (19.9%) experienced symptoms related to cardiotoxicity during therapy, which accumulated among those with EF drop. Underlying cardiovascular diseases and multiple (≥ 2) cardiac medications were related to EF drop (≥ 20%) and trastuzumab discontinuation. Majority of EF drops (≥ 10%) and trastuzumab discontinuations were seen within 6months of trastuzumab initiation and recovery of EF drop to < 10% of the baseline was seen in most cases (62.5%). There was no statistically significant difference in the survival of patients according to EF drop. CONCLUSIONS: Trastuzumab cardiotoxicity seems to accumulate among patients with underlying cardiac conditions. EF monitoring could be targeted to risk groups without compromising of the cardiac health or survival of HER2-positive breast cancer patients.


Assuntos
Antineoplásicos Imunológicos/efeitos adversos , Neoplasias da Mama/tratamento farmacológico , Trastuzumab/efeitos adversos , Adulto , Idoso , Idoso de 80 Anos ou mais , Cardiotoxicidade/epidemiologia , Cardiotoxicidade/etiologia , Feminino , Humanos , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Risco
6.
Duodecim ; 133(6): 599-605, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29243892

RESUMO

Clinical development of a novel drug has traditionally been seen as a series of four phases, each having its own objectives in establishing the efficacy and safety of the drug. Increasingly individualized medicine and the changing mechanisms of drug action are also changing the designs of clinical drug testing. The borders of development phases become blurred and the traditional large, controlled multicenter studies may in part be replaced by individual and risk-based approaches. The indications for drugs are more precisely targeted from biological starting points, and a target-oriented development may guide the designs of clinical testing at all stages of development. Utilization of data from registries along with modeling will become more common in clinical drug testing.


Assuntos
Ensaios Clínicos como Assunto , Avaliação de Medicamentos , Projetos de Pesquisa , Humanos , Medicina de Precisão , Sistema de Registros
7.
Eur J Heart Fail ; 18(2): 133-41, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26470631

RESUMO

In the past decade, novel cell-based products have been studied in patients with acute and chronic cardiac disease to assess whether these therapies are efficacious in improving heart function and preventing the development of end-stage heart failure. Cardiac indications studied include acute myocardial infarction (AMI), refractory angina, and chronic heart failure (CHF). Increased clinical activity, experience, and multiple challenges faced by developers have been recognized at the regulatory level. In May 2014, the Committee for Advanced Therapies (CAT) discussed in an expert meeting various cell-based medicinal products developed for cardiac repair, with a focus on non-manipulated bone marrow cells, sorted bone marrow or apheresis, and expanded cells, applied to patients with AMI or CHF. The intention was to share information, both scientific and regulatory, and to examine the challenges and opportunities in this field. These aspects were considered from the quality, and non-clinical and clinical perspectives, including current imaging techniques, with a focus on AMI and CHF. The scope of this overview is to present the European regulatory viewpoint on cell-based therapies for cardiac repair in the context of scientific observations.


Assuntos
Transplante de Células/métodos , Insuficiência Cardíaca/terapia , Coração/fisiologia , Infarto do Miocárdio/terapia , Regeneração , Animais , Modelos Animais de Doenças , Europa (Continente) , Insuficiência Cardíaca/diagnóstico , Humanos , Infarto do Miocárdio/diagnóstico
8.
Duodecim ; 131(5): 441-7, 2015.
Artigo em Finlandês | MEDLINE | ID: mdl-26237906

RESUMO

With the development of new protein kinases, antibodies, immunomodulators and hormonal treatments, significant progress has taken place in the last few years in the pharmacological treatment of cancers. At the same time, old cytotoxic drugs still remain in use. In regard to the heart, anthracyclines are the most problematic cytotoxic drugs, as they may cause cardiac insufficiency that is manifested only years after the treatment. Also trastuzumab and kinase inhibitors may cause cardiac insufficiency. Fluoropyrimidines cause myocardial ischemia for some patients. Treatments targeting VEGF inhibition are frequently associated with significant elevation of blood pressure.


Assuntos
Antineoplásicos/efeitos adversos , Cardiopatias/induzido quimicamente , Coração/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Antraciclinas/efeitos adversos , Anticorpos Monoclonais Humanizados/efeitos adversos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Fluoruracila/efeitos adversos , Humanos , Inibidores de Proteínas Quinases/efeitos adversos , Fatores de Risco , Trastuzumab , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
9.
PLoS One ; 10(6): e0130502, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26098115

RESUMO

The phosphatase and actin regulator 1 (PHACTR1) locus is a very commonly identified hit in genome-wide association studies investigating coronary artery disease and myocardial infarction (MI). However, the function of PHACTR1 in the heart is still unknown. We characterized the mechanisms regulating Phactr1 expression in the heart, used adenoviral gene delivery to investigate the effects of Phactr1 on cardiac function, and analyzed the relationship between MI associated PHACTR1 allele and cardiac function in human subjects. Phactr1 mRNA and protein levels were markedly reduced (60%, P<0.01 and 90%, P<0.001, respectively) at 1 day after MI in rats. When the direct myocardial effects of Phactr1 were studied, the skeletal α-actin to cardiac α-actin isoform ratio was significantly higher (1.5-fold, P<0.05) at 3 days but 40% lower (P<0.05) at 2 weeks after adenovirus-mediated Phactr1 gene delivery into the anterior wall of the left ventricle. Similarly, the skeletal α-actin to cardiac α-actin ratio was lower at 2 weeks in infarcted hearts overexpressing Phactr1. In cultured neonatal cardiac myocytes, adenovirus-mediated Phactr1 overexpression for 48 hours markedly increased the skeletal α-actin to cardiac α-actin ratio, this being associated with an enhanced DNA binding activity of serum response factor. Phactr1 overexpression exerted no major effects on the expression of other cardiac genes or LV structure and function in normal and infarcted hearts during 2 weeks' follow-up period. In human subjects, MI associated PHACTR1 allele was not associated significantly with cardiac function (n = 1550). Phactr1 seems to regulate the skeletal to cardiac α-actin isoform ratio.


Assuntos
Actinas/metabolismo , Cardiomiopatias/metabolismo , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Infarto do Miocárdio/metabolismo , Actinas/genética , Alelos , Animais , Cardiomiopatias/genética , Estudos de Casos e Controles , Células Cultivadas , Humanos , Masculino , Infarto do Miocárdio/genética , Miócitos Cardíacos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
10.
PLoS One ; 10(4): e0124907, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25915632

RESUMO

AIMS: In a recent genome-wide association study, WD-repeat domain 12 (WDR12) was associated with early-onset myocardial infarction (MI). However, the function of WDR12 in the heart is unknown. METHODS AND RESULTS: We characterized cardiac expression of WDR12, used adenovirus-mediated WDR12 gene delivery to examine effects of WDR12 on left ventricular (LV) remodeling, and analyzed relationship between MI associated WDR12 allele and cardiac function in human subjects. LV WDR12 protein levels were increased in patients with dilated cardiomyopathy and rats post-infarction. In normal adult rat hearts, WDR12 gene delivery into the anterior wall of the LV decreased interventricular septum diastolic and systolic thickness and increased the diastolic and systolic diameters of the LV. Moreover, LV ejection fraction (9.1%, P<0.05) and fractional shortening (12.2%, P<0.05) were declined. The adverse effects of WDR12 gene delivery on cardiac function were associated with decreased cellular proliferation, activation of p38 mitogen-activated protein kinase (MAPK)/heat shock protein (HSP) 27 pathway, and increased protein levels of Block of proliferation 1 (BOP1), essential for ribosome biogenesis. Post-infarction WDR12 gene delivery decreased E/A ratio (32%, P<0.05) suggesting worsening of diastolic function. In human subjects, MI associated WDR12 allele was associated significantly with diastolic dysfunction and left atrial size. CONCLUSIONS: WDR12 triggers distinct deterioration of cardiac function in adult rat heart and the MI associated WDR12 variant is associated with diastolic dysfunction in human subjects.


Assuntos
Insuficiência Cardíaca/metabolismo , Hemodinâmica , Infarto do Miocárdio/metabolismo , Proteínas Nucleares/metabolismo , Regulação para Cima , Adulto , Alelos , Animais , Proteínas de Ciclo Celular , Células Cultivadas , Feminino , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico HSP27/metabolismo , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/genética , Proteínas de Ligação a RNA , Ratos , Ratos Sprague-Dawley , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
PLoS One ; 9(4): e93473, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24695532

RESUMO

BACKGROUND: Apelin, the endogenous ligand for the G protein-coupled apelin receptor, is an important regulator of the cardiovascular homoeostasis. We previously demonstrated that apelin is one of the most potent endogenous stimulators of cardiac contractility; however, its underlying signaling mechanisms remain largely elusive. In this study we characterized the contribution of protein kinase C (PKC), extracellular signal-regulated kinase 1/2 (ERK1/2) and myosin light chain kinase (MLCK) to the positive inotropic effect of apelin. METHODS AND RESULTS: In isolated perfused rat hearts, apelin increased contractility in association with activation of prosurvival kinases PKC and ERK1/2. Apelin induced a transient increase in the translocation of PKCε, but not PKCα, from the cytosol to the particulate fraction, and a sustained increase in the phosphorylation of ERK1/2 in the left ventricle. Suppression of ERK1/2 activation diminished the apelin-induced increase in contractility. Although pharmacological inhibition of PKC attenuated the inotropic response to apelin, it had no effect on ERK1/2 phosphorylation. Moreover, the apelin-induced positive inotropic effect was significantly decreased by inhibition of MLCK, a kinase that increases myofilament Ca2+ sensitivity. CONCLUSIONS: Apelin increases cardiac contractility through parallel and independent activation of PKCε and ERK1/2 signaling in the adult rat heart. Additionally MLCK activation represents a downstream mechanism in apelin signaling. Our data suggest that, in addition to their role in cytoprotection, modest activation of PKCε and ERK1/2 signaling improve contractile function, therefore these pathways represent attractive possible targets in the treatment of heart failure.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Contração Muscular/fisiologia , Contração Miocárdica/fisiologia , Proteína Quinase C-épsilon/metabolismo , Animais , Apelina , Ventrículos do Coração/metabolismo , Ventrículos do Coração/fisiopatologia , Masculino , Quinase de Cadeia Leve de Miosina/metabolismo , Fosforilação/fisiologia , Proteína Quinase C-alfa/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia
12.
PLoS One ; 7(7): e41404, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22911790

RESUMO

BACKGROUND: Activation of the renin-angiotensin-system (RAS) plays a key pathophysiological role in heart failure in patients with hypertension and myocardial infarction. However, the function of (pro)renin receptor ((P)RR) is not yet solved. We determined here the direct functional and structural effects of (P)RR in the heart. METHODOLOGY/PRINCIPAL FINDINGS: (P)RR was overexpressed by using adenovirus-mediated gene delivery in normal adult rat hearts up to 2 weeks. (P)RR gene delivery into the anterior wall of the left ventricle decreased ejection fraction (P<0.01), fractional shortening (P<0.01), and intraventricular septum diastolic and systolic thickness, associated with approximately 2-fold increase in left ventricular (P)RR protein levels at 2 weeks. To test whether the worsening of cardiac function and structure by (P)RR gene overexpression was mediated by angiotensin II (Ang II), we infused an AT(1) receptor blocker losartan via osmotic minipumps. Remarkably, cardiac function deteriorated in losartan-treated (P)RR overexpressing animals as well. Intramyocardial (P)RR gene delivery also resulted in Ang II-independent activation of extracellular-signal-regulated kinase1/2 phosphorylation and myocardial fibrosis, and the expression of transforming growth factor-ß1 and connective tissue growth factor genes. In contrast, activation of heat shock protein 27 phosphorylation and apoptotic cell death by (P)RR gene delivery was Ang II-dependent. Finally, (P)RR overexpression significantly increased direct protein-protein interaction between (P)RR and promyelocytic zinc-finger protein. CONCLUSIONS/SIGNIFICANCE: These results indicate for the first time that (P)RR triggers distinct Ang II-independent myocardial fibrosis and deterioration of cardiac function in normal adult heart and identify (P)RR as a novel therapeutic target to optimize RAS blockade in failing hearts.


Assuntos
Matriz Extracelular/metabolismo , Testes de Função Cardíaca/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo , Adenoviridae/genética , Angiotensina II/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Fibrose , Técnicas de Transferência de Genes , Proteínas de Choque Térmico HSP27/metabolismo , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Ventrículos do Coração/fisiopatologia , Fatores de Transcrição Kruppel-Like/metabolismo , Losartan/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Especificidade de Órgãos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular/genética , Regulação para Cima/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos , Receptor de Pró-Renina
13.
Cardiovasc Res ; 89(1): 204-13, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20733007

RESUMO

AIMS: heart growth and function are angiogenesis-dependent, but little is known concerning the effects of key regulators of angiogenesis on diastolic heart failure. Here, we tested the hypothesis that local vascular endothelial growth factor-B (VEGF-B) gene therapy prevents left ventricular diastolic dysfunction. METHODS AND RESULTS: rats were subjected to pressure overload by infusing angiotensin II (33.3 microg/kg/h) for 2 weeks using osmotic minipumps. Intramyocardial delivery of adenoviral vector expressing VEGF-B(167A) improved the angiotensin II-induced diastolic dysfunction compared with LacZ control virus. Local VEGF-B gene transfer increased the mean capillary area in the left ventricle in control and angiotensin II-infused animals, whereas the density of capillaries was not affected. Interestingly, significant increases were noted in Ki67(+) proliferating cells, expression of interleukin1ß, and c-kit(+) cells in response to VEGF-B gene transfer. The increase in cardiac c-kit(+) cells was not associated with an induction of stromal cell-derived factor 1α, suggesting no mobilization of cells from bone marrow. Also, the phosphatidylinositol 3-kinase/Akt pathway was activated. CONCLUSION: VEGF-B gene transfer resulted in prevention of the angiotensin II-induced diastolic dysfunction associated with induction of the Akt pathway, increased proliferation and number of c-kit(+) cells, as well as an increase in the capillary area in the left ventricle. VEGF-B may offer novel therapeutic possibilities for the prevention of the transition from compensated to decompensated cardiac hypertrophy and thereby for the treatment of heart failure.


Assuntos
Insuficiência Cardíaca Diastólica/prevenção & controle , Fator B de Crescimento do Endotélio Vascular/genética , Adenoviridae/genética , Angiotensina II/administração & dosagem , Animais , Animais Geneticamente Modificados , Apoptose , Capilares/patologia , Proliferação de Células , Modelos Animais de Doenças , Fibrose , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos , Insuficiência Cardíaca Diastólica/etiologia , Insuficiência Cardíaca Diastólica/patologia , Insuficiência Cardíaca Diastólica/fisiopatologia , Humanos , Miócitos Cardíacos/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapêutico , Fator B de Crescimento do Endotélio Vascular/fisiologia , Fator B de Crescimento do Endotélio Vascular/uso terapêutico
14.
Circ Heart Fail ; 3(3): 440-50, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20200331

RESUMO

BACKGROUND: Recent data suggest that GATA-4 is an antiapoptotic factor required for adaptive responses and a key regulator of hypertrophy and hypertrophy-associated genes in the heart. As a leading cause of chronic heart failure, reversal of postinfarction left ventricular remodeling represents an important target for therapeutic interventions. Here, we studied the role of GATA-4 as a mediator of postinfarction remodeling in rats. METHODS AND RESULTS: Myocardial infarction, caused by ligating the left anterior descending coronary artery, significantly decreased the DNA binding activity of GATA-4 at day 1, whereas at 2 weeks the GATA-4 DNA binding was significantly upregulated. To determine the functional role of GATA-4, peri-infarct intramyocardial delivery of adenoviral vector expressing GATA-4 was done before left anterior descending coronary artery ligation. Hearts treated with GATA-4 gene transfer exhibited significantly increased ejection fraction and fractional shortening. Accordingly, infarct size was significantly reduced. To determine the cardioprotective mechanisms of GATA-4, myocardial angiogenesis, rate of apoptosis, c-kit+ cardiac stemlike cells, and genes regulated by GATA-4 were studied. The number of capillaries and stemlike cells was significantly increased, and decreased apoptosis was observed. CONCLUSION: These results indicate that the reversal of reduced GATA-4 activity prevents adverse postinfarction remodeling through myocardial angiogenesis, antiapoptosis, and stem cell recruitment. GATA-4-based gene transfer may represent a novel, efficient therapeutic approach for heart failure.


Assuntos
Fator de Transcrição GATA4/fisiologia , Insuficiência Cardíaca/prevenção & controle , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Neovascularização Fisiológica/fisiologia , Remodelação Ventricular/fisiologia , Animais , Apoptose , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/patologia , Masculino , Infarto do Miocárdio/terapia , Ratos , Ratos Sprague-Dawley
15.
Blood Press ; 19(1): 54-63, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20175653

RESUMO

Dyxin is a novel LIM domain protein acting as a transcriptional cofactor with GATA transcription factors. Here, we characterized dyxin as a p38 mitogen-activated protein kinase (MAPK) regulated gene, since combined upstream MAPK kinase 3b and wild-type p38 alpha MAPK gene transfer increased left ventricular dyxin mRNA and protein levels in vivo. We also studied cardiac dyxin expression in experimental models of pressure overload and myocardial infarction (MI) in vivo. Angiotensin II infusion increased left ventricular dyxin mRNA levels (9.4-fold, p<0.001) rapidly at 6 h followed by induction of protein levels. Furthermore, simultaneous administration of p38 MAPK inhibitor SB203580 abolished angiotensin II-induced activation of dyxin gene expression. During the post-infarction remodeling process, increased dyxin mRNA levels (7.7-fold, p<0.01) were noted at day 1 followed by the increase in proteins levels at 2 weeks after MI (1.5-fold, p<0.05). Moreover, direct wall stretch by using isolated rat heart preparation as well as direct mechanical stretch of cardiomyocytes in vitro activated dyxin gene expression within 1 h. Our results indicate that dyxin expression is rapidly upregulated in response to mechanical load, this increase being at least partly mediated by p38 MAPK. These results suggest that dyxin may play an important role in regulating hypertrophic process.


Assuntos
Proteínas de Transporte/metabolismo , Coração/fisiopatologia , Miocárdio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Adenoviridae/genética , Angiotensina II , Animais , Proteínas de Transporte/genética , Proteínas Correpressoras , Técnicas de Transferência de Genes , Vetores Genéticos , Ventrículos do Coração/metabolismo , Hipertensão/induzido quimicamente , Hipertensão/fisiopatologia , Técnicas In Vitro , Proteínas com Domínio LIM , Masculino , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Estresse Mecânico , Vasoconstritores , Remodelação Ventricular
16.
Clin Transl Sci ; 2(6): 422-30, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20443934

RESUMO

There is strong evidence for the use of angiotensin converting enzyme inhibitors and beta-blockers to reduce morbidity and mortality in patients with myocardial infarction (MI), whereas the effect of angiotensin receptor blockers is less clear. We evaluated the effects of an angiotensin receptor blocker losartan and a beta-blocker metoprolol on left ventricular (LV) remodeling, c-kit+ cells, proliferation, fibrosis, apoptosis, and angiogenesis using a model of coronary ligation in rats. Metoprolol treatment for 2 weeks improved LV systolic function. In contrast, losartan triggered deleterious structural remodeling and functional deterioration of LV systolic function, ejection fraction being 41% and fractional shortening 47% lower in losartan group than in controls 2 weeks after MI. The number of c-kit+ cells as well as expression of Ki-67 was increased by metoprolol. Losartan-induced thinning of the anterior wall and ventricular dilation were associated with increased apoptosis and fibrosis, while losartan had no effect on the expression of c-kit or Ki-67. Metoprolol or losartan had no effect on microvessel density. These results demonstrate that beta-blocker treatment attenuated adverse remodeling via c-kit+ cells and proliferation, whereas angiotensin receptor blocker-induced worsening of LV systolic function was associated with increased apoptosis and fibrosis in the peri-infarct region.


Assuntos
Apoptose/efeitos dos fármacos , Ventrículos do Coração/fisiopatologia , Losartan/farmacologia , Metoprolol/farmacologia , Infarto do Miocárdio/fisiopatologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Remodelação Ventricular/efeitos dos fármacos , Animais , Anti-Hipertensivos/farmacologia , Anti-Hipertensivos/uso terapêutico , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Interleucina-1beta/metabolismo , Losartan/uso terapêutico , Metoprolol/uso terapêutico , Infarto do Miocárdio/complicações , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Neovascularização Patológica/complicações , Neovascularização Patológica/fisiopatologia , Ratos , Função Ventricular Esquerda/efeitos dos fármacos
17.
FASEB J ; 20(11): 1907-9, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16849392

RESUMO

As a leading cause of heart failure, postinfarction left ventricular remodeling represents an important target for therapeutic interventions. Mitogen-activated protein kinases regulate critical cellular processes including stress response and survival, but their role in left ventricular remodeling is unknown. In the present study, rats were subjected to myocardial infarction by ligating the left anterior descending coronary artery. Western blot and kinase assay analysis revealed an inactivation of p38 kinase after myocardial infarction. Local adenovirus-mediated cotransfection of wild-type (WT) p38 kinase and constitutively active MKK3b reduced infarct size (26+/-3% vs. 47+/-4%, P<0.05 vs. LacZ-treated control) associated with improved ejection fraction (66.9+/-5.5% vs. 44.4+/-4.0%, P<0.001), fractional shortening (30.2+/-2.1% vs. 19.7+/-2.2%, P<0.001), and decreased left ventricular diastolic diameter (8.5+/-0.4 mm vs. 9.5+/-0.2 mm, P<0.01). p38 kinase gene transfer increased capillary density (2423+/-107/mm(2) vs. 1934+/-86/mm(2), P<0.001) and resulted in microvessel enlargement in the ischemic border zone. Apoptosis (35+/-7 vs. 69+/-13 cells, P<0.01) and fibrosis (16+/-3% vs. 34+/-8%, P<0.05) were reduced, while the number of c-kit positive cardiac stem-like cells remained unchanged. These results indicate that reduced p38 signaling predisposes to adverse postinfarction remodeling. The rescue of failing myocardium with p38 kinase may be a potential new therapy for heart failure after myocardial infarction.


Assuntos
Indutores da Angiogênese/uso terapêutico , Apoptose/efeitos dos fármacos , Insuficiência Cardíaca/terapia , Infarto do Miocárdio/terapia , Remodelação Ventricular , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Adenoviridae , Animais , Modelos Animais de Doenças , Eletrocardiografia , Ativação Enzimática , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos , Insuficiência Cardíaca/etiologia , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Infarto do Miocárdio/complicações , Ratos , Ratos Sprague-Dawley
18.
Circ Res ; 99(5): 485-93, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16873723

RESUMO

Mitogen-activated protein kinases (MAPKs) regulate cardiomyocyte growth and apoptosis in response to extracellular stimulation, but the downstream effectors that mediate their pathophysiological effects remain poorly understood. We determined the targets and role of p38 MAPK in the heart in vivo by using local adenovirus-mediated gene transfer of constitutively active upstream kinase mitogen-activated protein kinase kinase 3b (MKK3bE) and wild-type p38alpha in rats. DNA microarray analysis of animals with cardiac-specific overexpression of p38 MAPK revealed that 264 genes were upregulated more than 2-fold including multiple genes controlling cell division, cell signaling, inflammation, adhesion, and transcription. A large number of previously unknown p38 target genes were found. Using gel mobility-shift assays we identified several cardiac transcription factors that were directly activated by p38 MAPK. Finally, we determined the functional significance of the altered cardiac gene-expression profile by histological analysis and echocardiographic measurements, which indicated that p38 MAPK overexpression-induced gene expression results in myocardial cell proliferation, inflammation, and fibrosis. In conclusion, we defined the novel target genes and transcription factors as well as the functional effects of p38 MAPK in the heart. Expression profiling of p38 MAPK overexpression identified cell cycle regulatory and inflammatory genes critical for pathological processes in the adult heart.


Assuntos
Ciclo Celular/genética , Regulação Enzimológica da Expressão Gênica , Inflamação/genética , Miocárdio/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Adenoviridae/genética , Animais , Proliferação de Células , Ecocardiografia , Perfilação da Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Isoenzimas/genética , MAP Quinase Quinase 3/genética , Miocárdio/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Fatores de Transcrição/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Regul Pept ; 128(3): 187-96, 2005 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-15837527

RESUMO

To study the molecular mechanisms for load-induced activation of BNP gene expression, increased wall stress was imposed on isolated isovolumetrically beating adult rat hearts by distension of a fluid filled balloon within the left ventricle. The wall stress for 2 h resulted in a 1.6-fold increase in the expression of BNP gene and a 2.0-fold increase of the c-fos gene. The inhibition of transcription by actinomycin D significantly decreased the baseline BNP mRNA levels but the wall stretch-induced increase of the gene expression remained unaffected. In contrast, the protein synthesis inhibitor cycloheximide increased baseline BNP mRNA levels and abolished the load-induced activation of gene expression. Furthermore, we studied the effects of protein kinase C (PKC), calcineurin and protein phosphatase 2A (PP2A) inhibition to characterize the role of intracellular pathways in the stretch-induced gene expression in the left ventricle. The expression of BNP and c-fos genes were not influenced by calcineurin, PP2A and PKC inhibition. In conclusion, we showed that the stretch-induced activation of BNP gene expression by increased left ventricular wall stress is independent of transcriptional mechanisms and dependent on protein synthesis. Moreover, our results suggest that the load-induced activation of BNP gene expression is independent of calcineurin, PKC and PP2A.


Assuntos
Calcineurina/fisiologia , Regulação da Expressão Gênica , Peptídeo Natriurético Encefálico/genética , Proteína Quinase C/metabolismo , Processamento Pós-Transcricional do RNA , Função Ventricular Esquerda/fisiologia , Animais , Cicloeximida/farmacologia , Dactinomicina/farmacologia , Hemodinâmica/efeitos dos fármacos , Masculino , Modelos Animais , Fosfoproteínas Fosfatases/metabolismo , Proteína Fosfatase 2 , Ratos , Ratos Sprague-Dawley , Estresse Mecânico
20.
J Biol Chem ; 279(23): 24852-60, 2004 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-15051723

RESUMO

The zinc finger transcription factor GATA-4 has been implicated as a critical regulator of inducible cardiac gene expression and as a potential mediator of the hypertrophic program. However, the precise intracellular mechanisms that regulate the DNA-binding activity of GATA-4 are not fully understood. The aim of the present study was to examine the role of mitogen-activated protein kinases (p38 kinase, extracellular signal-regulated protein kinase, and c-Jun N-terminal protein kinase) in the left ventricular wall stress-induced activation of GATA-4 DNA binding in adult heart. Isolated perfused rat hearts were subjected to increased left ventricular wall stress by inflating a balloon in the ventricle. Gel mobility shift assays were used to analyze the transacting factors that interact with the GATA motifs of the B-type natriuretic peptide promoter. The left ventricular wall stress rapidly activated GATA-4 DNA binding and significantly increased the levels of phosphorylated p38 kinase, extracellular signal-regulated protein kinase, and c-Jun N-terminal protein kinase. The wall stress-induced increase in the DNA-binding activity of GATA-4 was abolished both in the presence of the p38 inhibitor SB239063 and MEK1/2 inhibitor U0126. In contrast, the inhibition of c-Jun N-terminal protein kinase by CEP11004 had no effect on the baseline or stretch-induced GATA-4 DNA binding. Moreover, GATA-4 DNA binding was up-regulated by mechanical stretch in the isolated rat atria via p38 and extracellular signal-regulated protein kinase. In conclusion, the present study demonstrates that both p38 and extracellular signal-regulated protein kinase are required for the stretch-induced GATA-4 binding in intact heart.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miocárdio/metabolismo , Fatores de Transcrição/metabolismo , Motivos de Aminoácidos , Animais , Northern Blotting , Western Blotting , Butadienos/farmacologia , Núcleo Celular/metabolismo , DNA/metabolismo , Endotelina-1/metabolismo , Inibidores Enzimáticos/farmacologia , Fator de Transcrição GATA4 , Ventrículos do Coração/enzimologia , Masculino , Proteína Quinase 3 Ativada por Mitógeno , Peptídeo Natriurético Encefálico/genética , Nitrilas/farmacologia , Perfusão , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Receptores de Angiotensina/metabolismo , Estresse Fisiológico , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...