Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 17(1): e0256512, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34995278

RESUMO

The mouse is a useful preclinical species for evaluating disease etiology due to the availability of a wide variety of genetically modified strains and the ability to perform disease-modifying manipulations. In order to establish an atrial filtration (AF) model in our laboratory, we profiled several commonly used murine AF models. We initially evaluated a pharmacological model of acute carbachol (CCh) treatment plus atrial burst pacing in C57BL/6 mice. In an effort to observe micro-reentrant circuits indicative of authentic AF, we employed optical mapping imaging in isolated mouse hearts. While CCh reduced atrial refractoriness and increased atrial tachyarrhythmia vulnerability, the left atrial (LA) excitation patterns were rather regular without reentrant circuits or wavelets. Therefore, the atrial tachyarrhythmia resembled high frequency atrial flutter, not typical AF per se. We next examined both a chronic angiotensin II (Ang II) infusion model and the surgical model of transverse aortic constriction (TAC), which have both been reported to induce atrial and ventricular structural changes that serve as a substrates for micro-reentrant AF. Although we observed some extent of atrial remodeling such as fibrosis or enlarged LA diameter, burst pacing-induced atrial tachyarrhythmia vulnerability did not differ from control mice in either model. This again suggested that an AF-like pathophysiology is difficult to demonstrate in the mouse. To continue searching for a valid murine AF model, we studied mice with a cardiac-specific deficiency (KO) in liver kinase B1 (Cardiac-LKB1), which has been reported to exhibit spontaneous AF. Indeed, the electrocardiograms (ECG) of conscious Cardiac-LKB1 KO mice exhibited no P waves and had irregular RR intervals, which are characteristics of AF. Histological evaluation of Cardiac-LKB1 KO mice revealed dilated and fibrotic atria, again consistent with AF. However, atrial electrograms and optical mapping revealed that electrical activity was limited to the sino-atrial node area with no electrical conduction into the atrial myocardium beyond. Thus, Cardiac-LKB1 KO mice have severe atrial myopathy or atrial standstill, but not AF. In summary, the atrial tachyarrhythmias we observed in the four murine models were distinct from typical human AF, which often exhibits micro- or macro-reentrant atrial circuits. Our results suggest that the four murine AF models we examined may not reflect human AF well, and raise a cautionary note for use of those murine models to study AF.


Assuntos
Fibrilação Atrial/fisiopatologia , Modelos Animais de Doenças , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Flutter Atrial/fisiopatologia , Função do Átrio Esquerdo/fisiologia , Remodelamento Atrial , Carbacol/farmacologia , Estimulação Cardíaca Artificial/efeitos adversos , Eletrocardiografia , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/patologia , Miócitos Cardíacos/patologia , Taquicardia Ventricular/fisiopatologia
3.
J Biol Chem ; 287(13): 9827-9834, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22228770

RESUMO

Insulin-like growth factor 1 (IGF1) promotes a physiological type of cardiac hypertrophy and has therapeutic effects in heart disease. Here, we report the relationship of IGF1 to GATA4, an essential transcription factor in cardiac hypertrophy and cell survival. In cultured neonatal rat ventricular myocytes, we compared the responses to IGF1 (10 nmol/liter) and phenylephrine (PE, 20 µmol/liter), a known GATA4 activator, in concentrations promoting a similar extent of hypertrophy. IGF1 and PE both increased nuclear accumulation of GATA4 and phosphorylation at Ser(105) (PE, 2.4-fold; IGF1, 1.8-fold; both, p < 0.05) and increased GATA4 DNA binding activity as indicated by ELISA and by chromatin IP of selected promoters. Although IGF1 and PE each activated GATA4 to the same degree, GATA4 knockdown by RNA interference only blocked hypertrophy by PE but not by IGF1. PE induction of a panel of GATA4 target genes (Nppa, Nppb, Tnni3, Myl1, and Acta1) was inhibited by GATA4 knockdown. In contrast, IGF1 regulated only Acta1 in a GATA4-dependent fashion. Consistent with the in vitro findings, Gata4 haploinsufficiency in mice did not alter cardiac structure, hyperdynamic function, or antifibrotic effects induced by myocardial overexpression of the IGF1 receptor. Our data indicate that GATA4 is activated by the IGF1 pathway, but although it is required for responses to pathological stimuli, it is not necessary for the effects of IGF1 on cardiac structure and function.


Assuntos
Cardiomegalia/metabolismo , Fator de Transcrição GATA4/metabolismo , Fator de Crescimento Insulin-Like I/efeitos adversos , Miócitos Cardíacos/metabolismo , Animais , Cardiomegalia/induzido quimicamente , Cardiomegalia/genética , Cardiomegalia/patologia , Células Cultivadas , Fator de Transcrição GATA4/genética , Técnicas de Silenciamento de Genes , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Fator de Crescimento Insulin-Like I/farmacologia , Camundongos , Camundongos Knockout , Miócitos Cardíacos/patologia , Regiões Promotoras Genéticas/genética , Ratos , Ratos Wistar
4.
Methods Mol Biol ; 573: 115-37, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19763925

RESUMO

Mouse models that mimic human diseases are important tools for investigating underlying mechanisms in many disease states. Although the demand for these models is high, there are few schools or courses available for surgeons to obtain the necessary skills. Researchers are usually exposed to brief descriptions of the procedures in scientific journals, which they then attempt to reproduce by trial and error. This often leads to a number of mistakes and unnecessary loss of animals. This chapter provides comprehensive details of three major surgical procedures currently employed in cardiovascular research: aortic constriction (of both ascending and transverse portions), pulmonary artery banding, and myocardial infarction (including ischemia-reperfusion). It guides the reader through the entire procedure, from the preparation of the animal for surgery until its full recovery, and includes a list of all necessary tools and devices. Due consideration has been given to the pitfalls and possible complications in the course of surgery. Adhering to our recommendations should improve reproducibility of the models and bring the number of the animal subjects to the minimum.


Assuntos
Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/cirurgia , Procedimentos Cirúrgicos Cardiovasculares/métodos , Modelos Animais de Doenças , Anestesia/métodos , Animais , Aorta/cirurgia , Pesquisa Biomédica/métodos , Pesquisa Biomédica/tendências , Procedimentos Cirúrgicos Cardiovasculares/instrumentação , Procedimentos Cirúrgicos Cardiovasculares/veterinária , Humanos , Camundongos , Modelos Biológicos , Infarto do Miocárdio/patologia , Infarto do Miocárdio/cirurgia , Cuidados Pós-Operatórios/métodos , Cuidados Pós-Operatórios/veterinária
5.
Nat Med ; 13(8): 952-61, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17660828

RESUMO

Cardiac fibrosis, associated with a decreased extent of microvasculature and with disruption of normal myocardial structures, results from excessive deposition of extracellular matrix, which is mediated by the recruitment of fibroblasts. The source of these fibroblasts is unclear and specific anti-fibrotic therapies are not currently available. Here we show that cardiac fibrosis is associated with the emergence of fibroblasts originating from endothelial cells, suggesting an endothelial-mesenchymal transition (EndMT) similar to events that occur during formation of the atrioventricular cushion in the embryonic heart. Transforming growth factor-beta1 (TGF-beta1) induced endothelial cells to undergo EndMT, whereas bone morphogenic protein 7 (BMP-7) preserved the endothelial phenotype. The systemic administration of recombinant human BMP-7 (rhBMP-7) significantly inhibited EndMT and the progression of cardiac fibrosis in mouse models of pressure overload and chronic allograft rejection. Our findings show that EndMT contributes to the progression of cardiac fibrosis and that rhBMP-7 can be used to inhibit EndMT and to intervene in the progression of chronic heart disease associated with fibrosis.


Assuntos
Diferenciação Celular , Fibrose Endomiocárdica/patologia , Células Endoteliais/patologia , Mesoderma/patologia , Animais , Células da Medula Óssea/patologia , Proteína Morfogenética Óssea 7 , Proteínas Morfogenéticas Ósseas/farmacologia , Proteínas Morfogenéticas Ósseas/uso terapêutico , Linhagem Celular , Células Cultivadas , Doença Crônica , Fibrose Endomiocárdica/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , Humanos , Células-Tronco Mesenquimais/patologia , Mesoderma/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta/uso terapêutico , Fator de Crescimento Transformador beta1/farmacologia
6.
Proc Natl Acad Sci U S A ; 103(39): 14471-6, 2006 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-16983087

RESUMO

An important event in the pathogenesis of heart failure is the development of pathological cardiac hypertrophy. In cultured cardiomyocytes, the transcription factor Gata4 is required for agonist-induced hypertrophy. We hypothesized that, in the intact organism, Gata4 is an important regulator of postnatal heart function and of the hypertrophic response of the heart to pathological stress. To test this hypothesis, we studied mice heterozygous for deletion of the second exon of Gata4 (G4D). At baseline, G4D mice had mild systolic and diastolic dysfunction associated with reduced heart weight and decreased cardiomyocyte number. After transverse aortic constriction (TAC), G4D mice developed overt heart failure and eccentric cardiac hypertrophy, associated with significantly increased fibrosis and cardiomyocyte apoptosis. Inhibition of apoptosis by overexpression of the insulin-like growth factor 1 receptor prevented TAC-induced heart failure in G4D mice. Unlike WT-TAC controls, G4D-TAC cardiomyocytes hypertrophied by increasing in length more than width. Gene expression profiling revealed up-regulation of genes associated with apoptosis and fibrosis, including members of the TGF-beta pathway. Our data demonstrate that Gata4 is essential for cardiac function in the postnatal heart. After pressure overload, Gata4 regulates the pattern of cardiomyocyte hypertrophy and protects the heart from load-induced failure.


Assuntos
Baixo Débito Cardíaco/induzido quimicamente , Baixo Débito Cardíaco/prevenção & controle , Fator de Transcrição GATA4/metabolismo , Coração/fisiologia , Pressão Ventricular/fisiologia , Animais , Aorta/fisiologia , Apoptose , Cardiomegalia/patologia , Células Cultivadas , Diástole/fisiologia , Fibrose , Fator de Transcrição GATA4/genética , Expressão Gênica , Regulação da Expressão Gênica , Coração/fisiopatologia , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor IGF Tipo 1/metabolismo , Sístole/fisiologia
7.
FASEB J ; 19(11): 1495-7, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15972800

RESUMO

Heterozygous mutations of the cardiac transcription factor Nkx2-5 cause congenital heart disease. To elucidate the molecular pathways of transcription factor mutant phenotypes or diseases, direct targets are commonly sought in studies of homozygous null mutant animals and by heterologous promoter-reporter gene transactivation assays. The expression of putative target genes in a physiologic range of transcription factor concentration, however, is often not examined. Heterozygous Nkx2-5 knockout (Nkx2-5+/-) mice have no more than half-normal levels of Nkx2-5 protein. We therefore measured the mRNA expression of four putative targets of the cardiac transcription factor Nkx2-5 in wild-type and Nkx2-5+/- animals in a variety of developmental and pathologic states. Wild-type and Nkx2-5+/- embryonic hearts expressed similar levels of atrial natriuretic factor (ANF), brain natriuretic peptide (BNP), the RNA helicase Csm, and homeodomain only protein HOP. In the failing adult ventricle, ANF and BNP were up-regulated to the same extent in wild-type and Nkx2-5+/- myocardium. Csm and HOP were down-regulated in heart failure, and Nkx2-5+/- hearts expressed about half-normal levels in healthy and failing states. No consistent relationship existed between the expression of putative transcriptional targets and Nkx2-5 gene dosage in the physiologically relevant range. Any dependence of gene expression on Nkx2-5 gene dosage is affected by factors specific to the individual gene and the physiologic context.


Assuntos
Regulação da Expressão Gênica , Proteínas de Homeodomínio/fisiologia , Miocárdio/metabolismo , Fatores de Transcrição/fisiologia , Animais , Fator Natriurético Atrial/genética , Conexinas/genética , Coração Fetal/metabolismo , Heterozigoto , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Masculino , Camundongos , Miocárdio/química , Peptídeo Natriurético Encefálico/genética , Fatores de Transcrição/genética , Proteína alfa-5 de Junções Comunicantes
8.
Mol Cell Biol ; 24(14): 6231-40, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15226426

RESUMO

Ribosomal S6 kinases (S6Ks) have been depicted as critical effectors downstream of growth factor pathways, which play an important role in the regulation of protein synthesis by phosphorylating the ribosomal protein, S6. The goal of this study was to determine whether S6Ks regulate heart size, are critical for the induction of cardiac hypertrophy in response to a pathological or physiological stimulus, and whether S6Ks are critical downstream effectors of the insulin-like growth factor 1 (IGF1)-phosphoinositide 3-kinase (PI3K) pathway. For this purpose, we generated and characterized cardiac-specific S6K1 and S6K2 transgenic mice and subjected S6K1(-/-), S6K2(-/-), and S6K1(-/-) S6K2(-/-) mice to a pathological stress (aortic banding) or a physiological stress (exercise training). To determine the genetic relationship between S6Ks and the IGF1-PI3K pathway, S6K transgenic and knockout mice were crossed with cardiac-specific transgenic mice overexpressing the IGF1 receptor (IGF1R) or PI3K mutants. Here we show that overexpression of S6K1 induced a modest degree of hypertrophy, whereas overexpression of S6K2 resulted in no obvious cardiac phenotype. Unexpectedly, deletion of S6K1 and S6K2 had no impact on the development of pathological, physiological, or IGF1R-PI3K-induced cardiac hypertrophy. These studies suggest that S6Ks alone are not essential for the development of cardiac hypertrophy.


Assuntos
Cardiomegalia/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptor IGF Tipo 1/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Transdução de Sinais/fisiologia , Animais , Antibióticos Antineoplásicos/metabolismo , Aorta/patologia , Feminino , Feto/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Tamanho do Órgão , Fosfatidilinositol 3-Quinases/genética , Condicionamento Físico Animal , Receptor IGF Tipo 1/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Sirolimo/metabolismo , Estresse Mecânico , Natação
9.
Circulation ; 109(24): 3050-5, 2004 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-15184287

RESUMO

BACKGROUND: Rapamycin is a specific inhibitor of the mammalian target of rapamycin (mTOR). We recently reported that administration of rapamycin before exposure to ascending aortic constriction significantly attenuated the load-induced increase in heart weight by approximately 70%. METHODS AND RESULTS: To examine whether rapamycin can regress established cardiac hypertrophy, mice were subjected to pressure overload (ascending aortic constriction) for 1 week, echocardiography was performed to verify an increase in ventricular wall thickness, and mice were given rapamycin (2 mg x kg(-1) x d(-1)) for 1 week. After 1 week of pressure overload (before treatment), 2 distinct groups of animals became apparent: (1) mice with compensated cardiac hypertrophy (normal function) and (2) mice with decompensated hypertrophy (dilated with depressed function). Rapamycin regressed the pressure overload-induced increase in heart weight/body weight (HW/BW) ratio by 68% in mice with compensated hypertrophy and 41% in mice with decompensated hypertrophy. Rapamycin improved left ventricular end-systolic dimensions, fractional shortening, and ejection fraction in mice with decompensated cardiac hypertrophy. Rapamycin also altered the expression of some fetal genes, reversing, in part, changes in alpha-myosin heavy chain and sarcoplasmic reticulum Ca2+ ATPase. CONCLUSIONS: Rapamycin may be a therapeutic tool to regress established cardiac hypertrophy and improve cardiac function.


Assuntos
Cardiomegalia/tratamento farmacológico , Proteínas Quinases/efeitos dos fármacos , Sirolimo/uso terapêutico , Adaptação Fisiológica , Animais , Aorta , Doenças da Aorta/complicações , Cardiomegalia/etiologia , Tamanho Celular/efeitos dos fármacos , Constrição Patológica/complicações , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Miócitos Cardíacos/patologia , Tamanho do Órgão/efeitos dos fármacos , Fosforilação , Proteínas Quinases/fisiologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteína S6 Ribossômica/metabolismo , Sirolimo/farmacologia , Volume Sistólico/efeitos dos fármacos , Serina-Treonina Quinases TOR
10.
Am J Physiol Heart Circ Physiol ; 287(1): H72-80, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15001440

RESUMO

Cardiac hypertrophy from pathological stimuli often proceeds to heart failure, whereas cardiac hypertrophy from physiological stimuli does not. In this study, physiological hypertrophy was created by a daily exercise regimen and pathological hypertrophy was created from a high-salt diet in Dahl salt-sensitive rats. The rats continued on a high-salt diet progressed to heart failure associated with an increased rate of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling-positive cardiomyocytes. We analyzed primary cultures of these hearts and found that only cardiomyocytes made hypertrophic by a pathological stimulus show increased sensitivity to apoptosis. Examination of the molecular changes associated with these distinct types of hypertrophy revealed changes in Bcl-2 family members and caspases favoring survival during physiological hypertrophy. However, in pathological hypertrophy, there were more diffuse proapoptotic changes, including changes in Fas, the Bcl-2 protein family, and caspases. Therefore, we speculate that this increased sensitivity to apoptotic stimulation along with proapoptotic changes in the apoptosis program may contribute to the development of heart failure seen in pathological cardiac hypertrophy.


Assuntos
Apoptose , Baixo Débito Cardíaco/fisiopatologia , Cardiomegalia/fisiopatologia , Animais , Baixo Débito Cardíaco/etiologia , Cardiomegalia/complicações , Cardiomegalia/etiologia , Caspases/genética , Caspases/metabolismo , Mitocôndrias Cardíacas , Atividade Motora , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos Dahl , Ratos Sprague-Dawley , Cloreto de Sódio na Dieta/administração & dosagem , Cloreto de Sódio na Dieta/efeitos adversos
11.
Physiol Genomics ; 16(3): 349-60, 2004 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-14679301

RESUMO

Mouse models mimicking human diseases are important tools in trying to understand the underlying mechanisms of many disease states. Several surgical models have been described that mimic human myocardial infarction (MI) and pressure-overload-induced cardiac hypertrophy. However, there are very few detailed descriptions for performing these surgical techniques in mice. Consequently, the number of laboratories that are proficient in performing cardiac surgical procedures in mice has been limited. Microarray technologies measure the expression of thousands of genes simultaneously, allowing for the identification of genes and pathways that may potentially be involved in the disease process. The statistical analysis of microarray experiments is highly influenced by the amount of variability in the experiment. To keep the number of required independent biological replicates and the associated costs of the study to a minimum, it is critical to minimize experimental variability by optimizing the surgical procedures. The aim of this publication was to provide a detailed description of techniques required to perform mouse cardiac surgery, such that these models can be utilized for genomic studies. A description of three major surgical procedures has been provided: 1) aortic constriction, 2) pulmonary artery banding, 3) MI (including ischemia-reperfusion). Emphasis has been placed on technical procedures with the inclusion of thorough descriptions of all equipment and devices employed in surgery, as well as the application of such techniques for expression profiling studies. The cardiac surgical techniques described have been, and will continue to be, important for elucidating the molecular mechanisms of cardiac hypertrophy and failure with high-throughput technology.


Assuntos
Cardiomegalia/genética , Modelos Animais de Doenças , Genômica/métodos , Genômica/normas , Infarto do Miocárdio/genética , Cirurgia Veterinária/métodos , Cirurgia Torácica/métodos , Anestesia/veterinária , Animais , Aorta/cirurgia , Cardiomegalia/etiologia , Cardiomegalia/patologia , Humanos , Intubação Intratraqueal/veterinária , Camundongos , Camundongos Endogâmicos , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Análise de Sequência com Séries de Oligonucleotídeos/normas , Cuidados Pós-Operatórios/veterinária , Artéria Pulmonar/cirurgia , Traumatismo por Reperfusão/veterinária , Reprodutibilidade dos Testes , Respiração Artificial/veterinária , Cirurgia Veterinária/normas , Taxa de Sobrevida , Cirurgia Torácica/normas , Pressão Ventricular
12.
J Biol Chem ; 279(6): 4782-93, 2004 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-14597618

RESUMO

Insulin-like growth factor 1 (IGF1) was considered a potential candidate for the treatment of heart failure. However, some animal studies and clinical trials have questioned whether elevating IGF1 chronically is beneficial. Secondary effects of increased serum IGF1 levels on other tissues may explain these unfavorable results. The aim of the current study was to examine the role of IGF1 in cardiac myocytes in the absence of secondary effects, and to elucidate downstream signaling pathways and transcriptional regulatory effects of the IGF1 receptor (IGF1R). Transgenic mice overexpressing IGF1R in the heart displayed cardiac hypertrophy, which was the result of an increase in myocyte size, and there was no evidence of histopathology. IGF1R transgenics also displayed enhanced systolic function at 3 months of age, and this was maintained at 12-16 months of age. The phosphoinositide 3-kinase (PI3K)-Akt-p70S6K1 pathway was significantly activated in hearts from IGF1R transgenics. Cardiac hypertrophy induced by overexpression of IGF1R was completely blocked by a dominant negative PI3K(p110alpha) mutant, suggesting IGF1R promotes compensated cardiac hypertrophy in a PI3K(p110alpha)-dependent manner. This study suggests that targeting the cardiac IGF1R-PI3K(p110alpha) pathway could be a potential therapeutic strategy for the treatment of heart failure.


Assuntos
Coração/crescimento & desenvolvimento , Fosfatidilinositol 3-Quinases/fisiologia , Receptor IGF Tipo 1/fisiologia , Animais , Sequência de Bases , Cardiomegalia/etiologia , DNA Complementar/genética , Coração/fisiologia , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Receptor IGF Tipo 1/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Sístole
13.
Proc Natl Acad Sci U S A ; 100(21): 12355-60, 2003 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-14507992

RESUMO

An unresolved question in cardiac biology is whether distinct signaling pathways are responsible for the development of pathological and physiological cardiac hypertrophy in the adult. Physiological hypertrophy is characterized by a normal organization of cardiac structure and normal or enhanced cardiac function, whereas pathological hypertrophy is associated with an altered pattern of cardiac gene expression, fibrosis, cardiac dysfunction, and increased morbidity and mortality. The elucidation of signaling cascades that play distinct roles in these two forms of hypertrophy will be critical for the development of more effective strategies to treat heart failure. We examined the role of the p110alpha isoform of phosphoinositide 3-kinase (PI3K) for the induction of pathological hypertrophy (pressure overload-induced) and physiological hypertrophy (exercise-induced) by using transgenic mice expressing a dominant negative (dn) PI3K(p110alpha) mutant specifically in the heart. dnPI3K transgenic mice displayed significant hypertrophy in response to pressure overload but not exercise training. dnPI3K transgenic mice also showed significant dilation and cardiac dysfunction in response to pressure overload. Thus, PI3K(p110alpha) appears to play a critical role for the induction of physiological cardiac growth but not pathological growth. PI3K(p110alpha) also appears essential for maintaining contractile function in response to pathological stimuli.


Assuntos
Cardiomegalia/enzimologia , Cardiomegalia/etiologia , Fosfatidilinositol 3-Quinases/fisiologia , Adaptação Fisiológica , Animais , Pressão Sanguínea , Cardiomegalia/genética , Tamanho Celular , Coração/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Modelos Cardiovasculares , Contração Miocárdica/fisiologia , Miócitos Cardíacos/patologia , Fenótipo , Fosfatidilinositol 3-Quinases/genética , Esforço Físico , Transdução de Sinais
14.
Circulation ; 107(12): 1664-70, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12668503

RESUMO

BACKGROUND: Cardiac hypertrophy, or an increase in heart size, is an important risk factor for cardiac morbidity and mortality. The mammalian target of rapamycin (mTOR) is a component of the insulin-phosphoinositide 3-kinase pathway, which is known to play a critical role in the determination of cell, organ, and body size. METHODS AND RESULTS: To examine the role of mTOR in load-induced cardiac hypertrophy, we administered rapamycin, a specific inhibitor of mTOR, to mice with ascending aortic constriction. Activity of p70 ribosomal S6 kinase 1 (S6K1), an effector of mTOR, was increased by 3.8-fold in the aortic-constricted heart. Pretreatment of mice with 2 mg. kg-1. d-1 of rapamycin completely suppressed S6K1 activation and S6 phosphorylation in response to pressure overload. The heart weight/tibial length ratio of vehicle-treated aortic-banded mice was increased by 34.4+/-3.6% compared with vehicle-treated sham-operated mice. Rapamycin suppressed the load-induced increase in heart weight by 67%. Attenuation of cardiac hypertrophy by rapamycin was associated with attenuation of the increase in myocyte cell size induced by aortic constriction. Rapamycin did not cause loss of body weight, lethality, or left ventricular dysfunction. CONCLUSIONS: mTOR or its target(s) seems to play an important role in load-induced cardiac hypertrophy. Because systemic administration of rapamycin has been used successfully for the treatment of transplant rejection in clinical practice, it may be a useful therapeutic modality to suppress cardiac hypertrophy in patients.


Assuntos
Cardiomegalia/etiologia , Inibidores de Proteínas Quinases , Sirolimo/farmacologia , Animais , Aorta , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Constrição , Feto/efeitos dos fármacos , Feto/metabolismo , Regulação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Coração/fisiopatologia , Hemodinâmica/efeitos dos fármacos , Masculino , Camundongos , Proteína Quinase 8 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/patologia , Tamanho do Órgão , Fosforilação , Proteínas Quinases/fisiologia , RNA Mensageiro/biossíntese , Proteína S6 Ribossômica/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Serina-Treonina Quinases TOR
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA