Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
PLoS One ; 14(6): e0218970, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31237922

RESUMO

We previously identified PEPCK-M (encoded by the Pck2 gene) to be highly up-regulated in skeletal muscle of pigs treated with Ractopamine, an anabolic beta-adrenergic receptor agonist. To determine whether PEPCK-M had a causative role in modulating the skeletal muscle growth response to Ractopamine, we used adeno-associated virus 1 (AAV1) to over-express Pck2 (AAV-Pck2) in murine skeletal muscle. A contralateral limb design was employed, such that each mouse served as its own control (injected with a GFP-only expressing AAV1, labelled AAV-GFP). Daily injections of Clenbuterol (1 mg/kg for 21 days) or vehicle control were also carried out to assess the effects of AAV-Pck2 overexpression on the anabolic response to a beta-adrenergic agonist. AAV-Pck2 overexpression in leg muscles of male C57BL6/J mice for 4 weeks (6-10 weeks of age) increased Pck2 mRNA (~100-fold), protein (not quantifiable) and enzyme activity (~3-fold). There was a trend (p = 0.0798) for AAV-Pck2 overexpression to reduce TA muscle weights, but there was no significant effect on muscle fibre diameters or myosin heavy chain isoform (MyHC) mRNA expression. When skeletal muscle growth was induced by daily administration of Clenbuterol (for 21 days), overexpression of AAV-Pck2 had no effect on the growth response, nor did it alter the expression of Phosphoserine Aminotransferase-1 (Psat1) or Asparagine Synthetase (Asns) mRNA or the Clenbuterol-induced decreases in MyHC IIa and IIx mRNA expression (p = 0.0065 and p = 0.0267 respectively). However AAV-Pck2 overexpression reduced TA muscle weights (p = 0.0434), particularly in the Control (vehicle treated) mice (p = 0.059 for AAV x Clenbuterol interaction) and increased the expression of Seryl-tRNA Synthetase (Sars) mRNA (p = 0.0477). Hence, contrary to the original hypothesis, AAV-Pck2 overexpression reduced TA muscle weights and did not mimic or alter the muscle hypertrophic effects of the beta-adrenergic agonist, Clenbuterol.


Assuntos
Clembuterol/farmacologia , Dependovirus/metabolismo , Desenvolvimento Muscular/efeitos dos fármacos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Fenetilaminas/farmacologia , Isoformas de Proteínas/metabolismo
2.
Eur Heart J ; 35(14): 932-41, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23990596

RESUMO

AIMS: Symptoms of cancer cachexia (CC) include fatigue, shortness of breath, and impaired exercise capacity, which are also hallmark symptoms of heart failure (HF). Herein, we evaluate the effects of drugs commonly used to treat HF (bisoprolol, imidapril, spironolactone) on development of cardiac wasting, HF, and death in the rat hepatoma CC model (AH-130). METHODS AND RESULTS: Tumour-bearing rats showed a progressive loss of body weight and left-ventricular (LV) mass that was associated with a progressive deterioration in cardiac function. Strikingly, bisoprolol and spironolactone significantly reduced wasting of LV mass, attenuated cardiac dysfunction, and improved survival. In contrast, imidapril had no beneficial effect. Several key anabolic and catabolic pathways were dysregulated in the cachectic hearts and, in addition, we found enhanced fibrosis that was corrected by treatment with spironolactone. Finally, we found cardiac wasting and fibrotic remodelling in patients who died as a result of CC. In living cancer patients, with and without cachexia, serum levels of brain natriuretic peptide and aldosterone were elevated. CONCLUSION: Systemic effects of tumours lead not only to CC but also to cardiac wasting, associated with LV-dysfunction, fibrotic remodelling, and increased mortality. These adverse effects of the tumour on the heart and on survival can be mitigated by treatment with either the ß-blocker bisoprolol or the aldosterone antagonist spironolactone. We suggest that clinical trials employing these agents be considered to attempt to limit this devastating complication of cancer.


Assuntos
Caquexia/prevenção & controle , Insuficiência Cardíaca/prevenção & controle , Neoplasias Hepáticas/prevenção & controle , Síndrome de Emaciação/prevenção & controle , Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Bisoprolol/farmacologia , Composição Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Imidazolidinas/farmacologia , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Cadeias Pesadas de Miosina/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos , Espironolactona/farmacologia , Análise de Sobrevida , Disfunção Ventricular Esquerda/tratamento farmacológico
3.
Oral Dis ; 19(7): 702-11, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23294197

RESUMO

OBJECTIVES: Chronic intermittent hypoxia (CIH) is a frequent feature of OSAHS. The present study was designed to evaluate the effects of genistein and estrogen on genioglossus contractile and regeneration properties in CIH rats and investigate the involvement of HIF-1α. METHODS: Ovariectomized female rats were exposed to CIH for 5 weeks. Genistein and estrogen were administered by intraperitoneal injection. The genioglossus myoblasts of rat were also isolated and cultured in vitro, and the HIF-1α shRNA lentivirus was used. RESULTS: Muscle fatigue resistance and myogenic regeneration were significantly decreased after CIH but were partially reversed by estrogen and genistein treatment. The effect of estrogen was more powerful than that of genistein. Compared with control group, RT-PCR and western blotting showed higher levels of HIF-1α mRNA and protein in the CIH group, but estrogen and genistein treatment reduced the levels of HIF-1α mRNA and protein in rats exposed to CIH. In genioglossus myoblasts, the expression of HIF-1α was up-regulated under hypoxia rather than normoxia and decreased over time under both hypoxia and normoxia during myogenic differentiation. HIF-1α knockdown relieved myogenesis inhibition under hypoxia. CONCLUSION: We concluded that genistein and estrogen may inhibit the overexpression of HIF-1α induced by CIH and improve the endurance and regeneration of the genioglossus muscle.


Assuntos
Estrogênios/farmacologia , Genisteína/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/efeitos dos fármacos , Músculos Faríngeos/efeitos dos fármacos , Fitoestrógenos/farmacologia , Apneia Obstrutiva do Sono/fisiopatologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Estrogênios/administração & dosagem , Feminino , Técnicas de Silenciamento de Genes , Inativação Gênica , Genisteína/administração & dosagem , Subunidade alfa do Fator 1 Induzível por Hipóxia/análise , Injeções Intraperitoneais , Lentivirus/genética , Contração Muscular/efeitos dos fármacos , Desenvolvimento Muscular/efeitos dos fármacos , Fadiga Muscular/efeitos dos fármacos , Proteína MyoD/efeitos dos fármacos , Mioblastos Esqueléticos/efeitos dos fármacos , Fator Regulador Miogênico 5/efeitos dos fármacos , Cadeias Pesadas de Miosina/efeitos dos fármacos , Ovariectomia , Fitoestrógenos/administração & dosagem , RNA Interferente Pequeno/genética , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Regeneração/efeitos dos fármacos
4.
Am J Physiol Cell Physiol ; 294(2): C613-26, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18003749

RESUMO

To understand the molecular mechanism of oxidation-induced inhibition of muscle contractility, we have studied the effects of hydrogen peroxide on permeabilized rabbit psoas muscle fibers, focusing on changes in myosin purified from these fibers. Oxidation by 5 mM peroxide decreased fiber contractility (isometric force and shortening velocity) without significant changes in the enzymatic activity of myofibrils and isolated myosin. The inhibitory effects were reversed by treating fibers with dithiothreitol. Oxidation by 50 mM peroxide had a more pronounced and irreversible inhibitory effect on fiber contractility and also affected enzymatic activity of myofibrils, myosin, and actomyosin. Peroxide treatment also affected regulation of contractility, resulting in fiber activation in the absence of calcium. Electron paramagnetic resonance of spin-labeled myosin in muscle fibers showed that oxidation increased the fraction of myosin heads in the strong-binding structural state under relaxing conditions (low calcium) but had no effect under activating conditions (high calcium). This change in the distribution of structural states of myosin provides a plausible explanation for the observed changes in both contractile and regulatory functions. Mass spectroscopy analysis showed that 50 mM but not 5 mM peroxide induced oxidative modifications in both isoforms of the essential light chains and in the heavy chain of myosin subfragment 1 by targeting multiple methionine residues. We conclude that 1) inhibition of muscle fiber contractility via oxidation of myosin occurs at high but not low concentrations of peroxide and 2) the inhibitory effects of oxidation suggest a critical and previously unknown role of methionines in myosin function.


Assuntos
Peróxido de Hidrogênio/metabolismo , Metionina/metabolismo , Contração Muscular/fisiologia , Músculo Esquelético/metabolismo , Miosinas/metabolismo , Actinas/metabolismo , Adenosina Trifosfatases/metabolismo , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Relação Dose-Resposta a Droga , Peróxido de Hidrogênio/farmacologia , Contração Isométrica/efeitos dos fármacos , Contração Isométrica/fisiologia , Espectrometria de Massas , Contração Muscular/efeitos dos fármacos , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/química , Músculo Esquelético/efeitos dos fármacos , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Miosinas/química , Miosinas/efeitos dos fármacos , Oxidantes/metabolismo , Oxidantes/farmacologia , Oxirredução/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Proteômica/métodos , Coelhos
5.
Prostate Cancer Prostatic Dis ; 9(4): 392-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16786038

RESUMO

PURPOSE: This study examined the effects of an 8-week androgen deprivation therapy treatment using Zoladex and an endurance training regimen on cardiac function. METHODS: Male Sprague-Dawley rats received either Zoladex or placebo. Animals remained sedentary or endurance trained during the drug treatment period. On day 57, ex vivo cardiac function was analyzed. RESULTS: Hearts from sedentary animals receiving Zoladex possessed significant cardiac dysfunction. However, hearts from exercise trained rats receiving Zoladex possessed cardiac function values similar to those from hearts from placebo animals. CONCLUSIONS: An 8-week treatment with Zoladex promoted cardiac dysfunction. Endurance training during Zoladex treatment protected against this cardiac dysfunction.


Assuntos
Antineoplásicos Hormonais/farmacologia , Circulação Coronária/efeitos dos fármacos , Resistência Física/efeitos dos fármacos , Pressão Ventricular/efeitos dos fármacos , Análise de Variância , Animais , Biomarcadores/sangue , Diástole/efeitos dos fármacos , Teste de Esforço , Gosserrelina/farmacologia , Coração/anatomia & histologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Ventrículos do Coração/fisiopatologia , Masculino , Cadeias Pesadas de Miosina/biossíntese , Cadeias Pesadas de Miosina/efeitos dos fármacos , Tamanho do Órgão , Ratos , Ratos Sprague-Dawley , Sístole/efeitos dos fármacos , Testículo/anatomia & histologia , Testosterona/sangue
6.
J Am Coll Cardiol ; 46(1): 125-33, 2005 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-15992646

RESUMO

OBJECTIVES: We assessed the effects of erythropoietin (EPO) treatment in a rat model of post-myocardial infarction (MI) heart failure. BACKGROUND: Erythropoietin, traditionally known as a hematopoietic hormone, has been linked to neovascularization. Whereas administration of EPO acutely after MI reduces infarct size and improves cardiac function, its role in the failing heart is unknown. METHODS: Rats underwent coronary ligation or sham surgery. Rats with MI were randomly assigned to: untreated (MI), a single bolus of EPO immediately after MI induction (MI-EPO-early), EPO treatment immediately after MI and once every three weeks (MI-EPO-early+late), and EPO treatment starting three weeks after induction of MI, once every three weeks (MI-EPO-late). After nine weeks, hemodynamics, infarct size, myosin heavy chain (MHC) isoforms, myocyte hypertrophy, and capillary density were measured. RESULTS: Erythropoietin treatment started immediately after MI (MI-EPO-early and MI-EPO-early+late) resulted in a 23% to 30% reduction in infarct size (p < 0.01) and, accordingly, hemodynamic improvement. Erythropoietin treatment, started three weeks after MI (MI-EPO-late), did not affect infarct size, but resulted in an improved cardiac performance, reflected by a 34% reduction in left ventricular end-diastolic pressure (p < 0.01), and 46% decrease in atrial natriuretic peptide levels (p < 0.05). The improved cardiac function was accompanied by an increased capillary density (p < 0.01), an increased capillary-to-myocyte ratio (p < 0.05), and a partial reversal of beta-MHC (p < 0.05) in all treated groups. CONCLUSIONS: In addition to its effect on infarct size reduction, EPO treatment improves cardiac function in a rat model of post-MI heart failure. This observation may be explained by neovascularization, associated with an increased alpha-MHC expression.


Assuntos
Débito Cardíaco/efeitos dos fármacos , Cardiomiopatia Dilatada/fisiopatologia , Vasos Coronários/efeitos dos fármacos , Eritropoetina/análogos & derivados , Eritropoetina/administração & dosagem , Infarto do Miocárdio/complicações , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Fator Natriurético Atrial/sangue , Fator Natriurético Atrial/efeitos dos fármacos , Cardiomiopatia Dilatada/etiologia , Cardiomiopatia Dilatada/metabolismo , Darbepoetina alfa , Modelos Animais de Doenças , Esquema de Medicação , Masculino , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Ratos , Ratos Sprague-Dawley
7.
Cardiovasc Drugs Ther ; 18(4): 257-67, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15367823

RESUMO

OBJECTIVE: Rapamycin inhibits p70 S6 kinase (p70(S6K)) activity and hypertrophy of cultured neonatal rat cardiac myocytes. The purpose of the present study was to determine whether rapamycin inhibits left ventricular (LV) hypertrophy in intact rats and whether it alters cardiac gene expression. METHODS: 300 g rats were subjected to aortic constriction (AC) or sham-operation (SH) and studied 2 and 3 days after surgery. Beginning 1 day prior to surgery, rats were injected with rapamycin (1.5 mg/kg, i.p.) or carboxymethylcellulose vehicle (V), yielding 4 groups (SH-V, SH-R, AC-V, AC-R). Total RNA was extracted for determination of mRNA levels by Northern blotting. RESULTS: LV dry weight/body weight ratios were 0.43 +/- 0.04 (mean +/- SE) for SH-V, 0.46 +/- 0.02 for SH-R, 0.56 +/- 0.02 for AC-V, and 0.53 +/- 0.03 for AC-R. R inhibited cardiac hypertrophy induced by pressure overload (ANOVA; p < 0.05). Rapamycin had no effect on the expression of atrial natriuretic factor mRNA, but increased the levels of beta-myosin heavy chain mRNA 6-fold in hearts of SH-R and AC-R compared to SH-V. Rapamycin also increased the expression of alpha-myosin heavy chain mRNA in SH-R by 3-fold compared with SH-V, but had no effect on the AC-R group. CONCLUSION: The data suggest that an intact mTOR signaling pathway is required for rapid hypertrophic growth of the heart in vivo. Moreover, the data suggest a novel link between the mTOR/p70(S6K) signal transduction pathway and pretranslational control of myosin gene expression in the heart.


Assuntos
Hipertrofia Ventricular Esquerda/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Proteínas Quinases/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Northern Blotting , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Hipertrofia Ventricular Esquerda/enzimologia , Imunossupressores/farmacologia , Masculino , Cadeias Pesadas de Miosina/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Quinases/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Quinases S6 Ribossômicas 70-kDa/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Regulação para Cima/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos
8.
J Clin Invest ; 114(3): 370-8, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15286803

RESUMO

Cachexia is a syndrome characterized by wasting of skeletal muscle and contributes to nearly one-third of all cancer deaths. Cytokines and tumor factors mediate wasting by suppressing muscle gene products, but exactly which products are targeted by these cachectic factors is not well understood. Because of their functional relevance to muscle architecture, such targets are presumed to represent myofibrillar proteins, but whether these proteins are regulated in a general or a selective manner is also unclear. Here we demonstrate, using in vitro and in vivo models of muscle wasting, that cachectic factors are remarkably selective in targeting myosin heavy chain. In myotubes and mouse muscles, TNF-alpha plus IFN-gamma strongly reduced myosin expression through an RNA-dependent mechanism. Likewise, colon-26 tumors in mice caused the selective reduction of this myofibrillar protein, and this reduction correlated with wasting. Under these conditions, however, loss of myosin was associated with the ubiquitin-dependent proteasome pathway, which suggests that mechanisms used to regulate the expression of muscle proteins may be cachectic factor specific. These results shed new light on cancer cachexia by revealing that wasting does not result from a general downregulation of muscle proteins but rather is highly selective as to which proteins are targeted during the wasting state.


Assuntos
Caquexia/metabolismo , Neoplasias do Colo/complicações , Regulação Neoplásica da Expressão Gênica , Músculo Esquelético/metabolismo , Proteína MyoD/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Animais , Células CHO , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Cricetinae , Cricetulus , Cisteína Endopeptidases/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Sinergismo Farmacológico , Marcação de Genes , Interferon gama/metabolismo , Interferon gama/farmacologia , Camundongos , Camundongos Endogâmicos , Camundongos Nus , Complexos Multienzimáticos/metabolismo , Músculo Esquelético/efeitos dos fármacos , Proteína MyoD/efeitos dos fármacos , Proteína MyoD/genética , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/genética , Complexo de Endopeptidases do Proteassoma , Sensibilidade e Especificidade , Transcrição Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Ubiquitinas/metabolismo
9.
Am J Physiol Heart Circ Physiol ; 286(2): H760-7, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14512277

RESUMO

The transient increase in the expression of transcription factors encoded by immediate-early genes has been considered to play a critical role in the coordination of early gene expression during the hypertrophic growth of cardiac myocytes. Here, we investigated the regulation of c-Jun and its upstream activators JNKs in the myocardium of rats subjected to acute pressure overload induced by transverse aortic constriction. Western blotting and immunohistochemistry analysis demonstrated that both JNK1 and JNK2 were transiently activated by pressure overload, but only JNK1 was activated at the nuclei of cardiac myocytes. JNK1 activation was paralleled by phosphorylation of c-Jun at serine-63 in the myocardial nuclear fraction and by an increase in c-Jun expression in cardiac myocytes. A consistent increase in DNA binding of activator protein-1 (AP-1) complex was observed after 10 and 30 min of pressure overload and Supershift assays confirmed that c-Jun was a major component of activated AP-1 complex. Moreover, experiments performed with the specific JNK inhibitor SP-600125 abolished c-Jun phosphorylation and markedly attenuated its expression as well as the expression of the fetal gene beta-myosin heavy chain. Overall, these findings demonstrate a molecular basis for load-induced activation of c-Jun in cardiac myocytes and its connection with the regulation of fetal gene, characteristic of the acute response to pressure overload.


Assuntos
Coração/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Animais , Antracenos/farmacologia , Sequência de Bases , Núcleo Celular/fisiologia , Citosol/fisiologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Coração/efeitos dos fármacos , Coração/fisiopatologia , Masculino , Proteína Quinase 8 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Modelos Animais , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/genética , Oligodesoxirribonucleotídeos , Fosforilação , Pressão , Proteínas Proto-Oncogênicas c-jun/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-jun/genética , Ratos , Ratos Wistar , Função Ventricular Esquerda/fisiologia
10.
Exp Cell Res ; 291(1): 56-69, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14597408

RESUMO

Inhibition of phosphatidylinositol 3-kinase (PI3-kinase) has been reported to block cardiomyocyte differentiation. However, at which stage PI3-kinase plays this important role and what its molecular targets are remain unknown. To answer these questions, we induced cardiomyocyte differentiation of P19CL6 mouse embryonal carcinoma cells and investigated the activation of PI3-kinase by analyzing phospho-Akt. We also treated P19CL6 cells with the PI3-kinase-specific inhibitor LY294002 either continuously or at various time points and monitored the expression of cardiac contractile proteins and transcription factors. Most cells differentiated into sarcomeric myosin heavy chain (MHC)-positive cardiomyocytes on day 16 after induction. An increase in phospho-Akt was observed after induction and was maintained throughout the differentiation. LY294002 treatment restricted to the phase from days 0 to 4 was sufficient to inhibit cardiomyocyte differentiation in a dose-dependent manner. In contrast, LY294002 treatment either from days 4 to 8 or from days 8 to 12 did not cause significant changes in sarcomeric MHC expression. LY294002 treatment from days 0 to 4 also suppressed Csx/Nkx-2.5 and GATA-4 expression. These results demonstrate that PI3-kinase becomes activated and plays a pivotal role at a very early stage of cardiomyocyte differentiation, possibly by modulating the expression of the cardiac transcription factors.


Assuntos
Diferenciação Celular/genética , Coração/embriologia , Miócitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Cromonas/farmacologia , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Esquema de Medicação , Inibidores Enzimáticos/farmacologia , Fator de Transcrição GATA4 , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/efeitos dos fármacos , Proteínas de Homeodomínio/metabolismo , Camundongos , Morfolinas/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/metabolismo
11.
J Gerontol A Biol Sci Med Sci ; 58(1): 3-10, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12560405

RESUMO

We tested the hypothesis that cross-bridge mechanisms of contraction differed in early postmenopausal women who did or did not receive hormone replacement therapy (HRT). Vastus lateralis biopsies were obtained from 17 postmenopausal women (49-57 years old), 8 of whom were on HRT for the previous 24 +/- 5 months and 9 of whom were never on HRT. Electrophoresis and enzyme histochemistry revealed that fiber myosin heavy chain (MHC) isoform distribution, the cross-sectional area (CSA) of slow and fast fibers, and the relative CSA occupied by each, were similar for HRT and non-HRT groups. Single permeabilized fibers containing type IIa MHC had greater Ca(2+)-activated peak specific force, unloaded shortening velocity, and peak power than fibers containing type I MHC, but in all cases the values for HRT and non-HRT groups were similar. In this cross-sectional study, we found no evidence that Ca(2+)-activated fiber function, MHC isoform distribution, or relative CSA occupied by slow and fast fibers differed between HRT and non-HRT groups.


Assuntos
Terapia de Reposição Hormonal , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/anatomia & histologia , Músculo Esquelético/fisiologia , Cadeias Pesadas de Miosina/efeitos dos fármacos , Pós-Menopausa/efeitos dos fármacos , Isoformas de Proteínas/efeitos dos fármacos , Antropometria , Biópsia por Agulha , Estudos de Casos e Controles , Técnicas de Cultura , Eletroforese em Gel de Ágar , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/fisiologia , Cadeias Pesadas de Miosina/metabolismo , Probabilidade , Isoformas de Proteínas/metabolismo , Valores de Referência , Estudos de Amostragem , Sensibilidade e Especificidade
12.
Am J Physiol Cell Physiol ; 284(3): C658-66, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12444016

RESUMO

The signal transduction pathways connecting cell surface receptors to the activation of muscle-specific promoters and leading to myogenesis are still largely unknown. Recently, a contribution of the p38 mitogen-activated protein kinase (MAPK) pathway to this process was evoked through the use of pharmacological inhibitors. We used several mutants of the kinases composing this pathway to modulate the activity of the muscle-specific myosin light chain and myogenin promoters in C2C12 cells by transient transfections. In addition, we show for the first time, using a stable C2C12 cell line expressing a dominant-negative form of the p38 activator MAPK kinase (MKK)3, that a functional p38 MAPK pathway is indeed required for terminal muscle cell differentiation. The most obvious phenotype of this cell line, besides the inhibition of the activation of p38, is its inability to undergo terminal differentiation. This phenotype is accompanied by a drastic inhibition of cell cycle and myogenesis markers such as p21, p27, MyoD, and troponin T, as well as a profound disorganization of the cytoskeleton.


Assuntos
Diferenciação Celular/genética , MAP Quinase Quinase Quinase 1 , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fibras Musculares Esqueléticas/enzimologia , Músculo Esquelético/embriologia , Músculo Esquelético/enzimologia , Mioblastos Esqueléticos/enzimologia , Proteínas Tirosina Quinases/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27 , MAP Quinase Quinase 3 , Camundongos , Proteína Quinase 14 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/genética , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Proteína MyoD/antagonistas & inibidores , Proteína MyoD/metabolismo , Mioblastos Esqueléticos/citologia , Miogenina/antagonistas & inibidores , Miogenina/metabolismo , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Cadeias Leves de Miosina/antagonistas & inibidores , Cadeias Leves de Miosina/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/efeitos dos fármacos , Proteínas Tirosina Quinases/genética , Proteínas Supressoras de Tumor/metabolismo
13.
J Card Fail ; 9(6): 469-74, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14966788

RESUMO

BACKGROUND: The left ventricular functional recovery by beta-blocker therapy is now attributed to time-dependent biologic effects on cardiomyocytes. METHODS AND RESULTS: To elucidate the cellular mechanism of these biologic effects, we treated 9 patients with dilated cardiomyopathy for 4 months with beta-blockers and examined the gene expressions linked to an improvement of left ventricular ejection fraction (EF). Gene expressions of the biopsied right ventricular endomyocardium were assessed by real-time reverse transcription-polymerase chain reaction. A decrease in beta-myosin heavy chain (1.23+/-0.49 versus 0.86+/-0.45, P<.05) was observed 4 months after the administration of beta-blockers. The expression levels of both sarcoplasmic reticulum Ca(2+) ATPase (SERCA) (0.80+/-0.28 versus 1.39+/-0.44, P<.01) and phospholamban (PLB) (0.49+/-0.08 versus 0.88+/-0.34, P<.05) increased, whereas the expression levels of Na(+)-Ca(2+) exchanger (NCX), beta-adrenoreceptor kinase 1, and ryanodine receptor 2 were unchanged. The SERCA/NCX ratio (0.68+/-0.14 versus 0.96+/-0.33, P<.05) also increased. The increase in SERCA mRNA expression correlated with the degree of changes in EF (%deltaEF) (r=0.679, P<.05), and none of changes in these genes expression correlated with changes in the plasma brain natriuretic peptide concentration. CONCLUSIONS: The functional recovery resulting from beta-blockers may be associated with the restoration of the unfavorable gene expression that controls Ca(2+) handlings in the failing heart.


Assuntos
Antagonistas Adrenérgicos beta/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Insuficiência Cardíaca/tratamento farmacológico , Miocárdio/química , Miocárdio/patologia , Adulto , Idoso , Biomarcadores/sangue , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/efeitos dos fármacos , ATPases Transportadoras de Cálcio/biossíntese , ATPases Transportadoras de Cálcio/efeitos dos fármacos , Cardiomiopatia Dilatada/complicações , Cardiomiopatia Dilatada/tratamento farmacológico , Cardiomiopatia Dilatada/genética , Proteínas Quinases Dependentes de AMP Cíclico/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Japão , Masculino , Pessoa de Meia-Idade , Contração Miocárdica/efeitos dos fármacos , Cadeias Pesadas de Miosina/biossíntese , Cadeias Pesadas de Miosina/efeitos dos fármacos , Peptídeo Natriurético Encefálico/sangue , Peptídeo Natriurético Encefálico/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Trocador de Sódio e Cálcio/efeitos dos fármacos , Trocador de Sódio e Cálcio/metabolismo , Volume Sistólico/efeitos dos fármacos , Resultado do Tratamento , Função Ventricular Esquerda/efeitos dos fármacos , Quinases de Receptores Adrenérgicos beta
14.
Nat Genet ; 32(4): 585-93, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12415271

RESUMO

Cell-cycle checkpoints help to protect the genomes of proliferating cells under genotoxic stress. In multicellular organisms, cell proliferation is often directed toward differentiation during development and throughout adult homeostasis. To prevent the formation of differentiated cells with genetic instability, we hypothesized that genotoxic stress may trigger a differentiation checkpoint. Here we show that exposure to genotoxic agents causes a reversible inhibition of myogenic differentiation. Muscle-specific gene expression is suppressed by DNA-damaging agents if applied prior to differentiation induction but not after the differentiation program is established. The myogenic determination factor, MyoD (encoded by Myod1), is a target of the differentiation checkpoint in myoblasts. The inhibition of MyoD by DNA damage requires a functional c-Abl tyrosine kinase (encoded by Abl1), but occurs in cells deficient for p53 (transformation-related protein 53, encoded by Trp53) or c-Jun (encoded by the oncogene Jun). These results support the idea that genotoxic stress can regulate differentiation, and identify a new biological function for DNA damage-activated signaling network.


Assuntos
Dano ao DNA , Mioblastos/metabolismo , Células 3T3 , Animais , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Cisplatino/farmacologia , Reparo do DNA , Etoposídeo/farmacologia , Metanossulfonato de Metila/farmacologia , Camundongos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Mutagênicos/farmacologia , Proteína MyoD/efeitos dos fármacos , Proteína MyoD/genética , Proteína MyoD/metabolismo , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Miogenina/efeitos dos fármacos , Miogenina/metabolismo , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Fosforilação , Mutação Puntual , Proteínas Proto-Oncogênicas c-abl/metabolismo , Proteínas Proto-Oncogênicas c-abl/fisiologia , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas c-jun/fisiologia , Radiação Ionizante , Ativação Transcricional/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/fisiologia , Tirosina/metabolismo
15.
Toxicology ; 179(3): 233-45, 2002 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12270595

RESUMO

The cadmium produced by industrial and agricultural practice represents a major environmental pollutant which may induce severe damage, especially in the kidney where cadmium accumulates. While cadmium is known to severely impair renal tubular functions, glomerular structures are also potential targets. The present study investigated the effects of cadmium on glomerular mesangial cell cultures after short- and long-term exposures, requiring for each endpoint specific culture conditions. After 30 min exposure to 1 microM CdCl(2), used as non-lethal concentration, 0.14 ng/microg proteins of cadmium was internalized by the cells as evaluated by atomic emision spectrometry and induced a significant, cell surface reduction (8.9+/-1.9%). These morphological changes could be correlated to smooth muscle alpha-actin disorganization, without quantitative change in its protein expression level as evaluated by Western-blot and Northern-blot analysis (SMAmRNA/28sRNA, 1.78 CdCl(2) vs. 1.42 control). For longer exposure times, in complex medium, cadmium uptake was efficient (0.36 ng/microg proteins) and induced changes in the actin cytoskeleton with no loss of cell membrane integrity. This study suggests that cultured mesangial cells provide an alternative model to study the effect of cadmium, and underlines the importance of using well-defined conditions to study further intracellular mechanisms.


Assuntos
Cádmio/toxicidade , Mesângio Glomerular/efeitos dos fármacos , Actinas/efeitos dos fármacos , Actinas/metabolismo , Animais , Cádmio/farmacocinética , Células Cultivadas , Relação Dose-Resposta a Droga , Mesângio Glomerular/fisiologia , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Masculino , Contração Muscular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Cadeias Leves de Miosina/efeitos dos fármacos , Cadeias Leves de Miosina/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Vimentina/efeitos dos fármacos , Vimentina/metabolismo
16.
Growth Horm IGF Res ; 12(4): 208-15, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12175653

RESUMO

OBJECTIVE: This study examined the effects of growth hormone (GH) on infarct size, survival, and cardiac gene expression in rats with acute myocardial infarction. DESIGN: Animals randomly received sc injection of either saline vehicle (n = 98) or GH (2mg/kg/day, n = 105) for 14 days commencing the day of left coronary artery ligation. Infarct size was determined by morphometric analysis at the time of death or at 52 weeks post-surgery. Gene expression was analyzed by real-time RT-PCR after 2-week treatment. RESULTS: GH decreased infarct size by 18% (P < 0.01) and increased survival by 36% at 52 weeks. GH also significantly reduced cardiac expression of atrial natriuretic factor, beta-myosin heavy chain, alpha-smooth muscle actin, collagen I, collagen III, fibronectin, and pro-inflammatory cytokines. CONCLUSIONS: Treatment with GH for 2 weeks beginning on the day of myocardial infarction produced beneficial effects that were associated with reductions in cardiac gene expression symptomatic of pathological remodeling.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio do Crescimento/farmacologia , Coração/fisiologia , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Actinas/efeitos dos fármacos , Actinas/genética , Animais , Fator Natriurético Atrial/efeitos dos fármacos , Fator Natriurético Atrial/genética , Peso Corporal/efeitos dos fármacos , Colágeno Tipo I/efeitos dos fármacos , Colágeno Tipo I/genética , Colágeno Tipo III/efeitos dos fármacos , Colágeno Tipo III/genética , Citocinas/efeitos dos fármacos , Citocinas/genética , Fibronectinas/efeitos dos fármacos , Fibronectinas/genética , Coração/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Masculino , Infarto do Miocárdio/genética , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/genética , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Taxa de Sobrevida
17.
N Engl J Med ; 346(18): 1357-65, 2002 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-11986409

RESUMO

BACKGROUND: Beta-blocker therapy may improve cardiac function in patients with idiopathic dilated cardiomyopathy. We tested the hypothesis that beta-blocker therapy produces favorable functional effects in dilated cardiomyopathy by altering the expression of myocardial genes that regulate contractility and pathologic hypertrophy. METHODS: We randomly assigned 53 patients with idiopathic dilated cardiomyopathy to treatment with a beta-adrenergic-receptor blocking agent (metoprolol or carvedilol) or placebo. The amount of messenger RNA (mRNA) for contractility-regulating genes (those encoding beta1- and beta2-adrenergic receptors, calcium ATPase in the sarcoplasmic reticulum, and alpha- and beta-myosin heavy-chain isoforms) and of genes associated with pathologic hypertrophy (beta-myosin heavy chain and atrial natriuretic peptide) was measured with a quantitative reverse-transcription polymerase chain reaction in total RNA extracted from biopsy specimens of the right ventricular septal endomyocardium. Myocardial levels of beta-adrenergic receptors were also measured. Measurements were conducted at base line and after six months of treatment, and changes in gene expression were compared with changes in the left ventricular ejection fraction as measured by radionuclide ventriculography. RESULTS: Twenty-six of 32 beta-blocker-treated patients (those with complete mRNA measurements) had an improvement in left ventricular ejection fraction of at least 5 ejection-fraction (EF) units (mean [+/-SE] increase, 18.8+/-1.8). As compared with the six beta-blocker-treated patients who did not have a response (mean change, a decrease of 2.5+/-1.8 EF units), those who did have a response had an increase in sarcoplasmic-reticulum calcium ATPase mRNA and alpha-myosin heavy chain mRNA and a decrease in beta-myosin heavy chain mRNA. The change in sarcoplasmic-reticulum calcium ATPase was not present in the patients in the placebo group who had a spontaneous response. There were no differences between those who had a response and those who did not in terms of the change in mRNA or protein expression of beta-adrenergic receptors. CONCLUSIONS: In idiopathic dilated cardiomyopathy, functional improvement related to treatment with beta-blockers is associated with changes in myocardial gene expression.


Assuntos
Antagonistas Adrenérgicos beta/uso terapêutico , Cardiomiopatia Dilatada/tratamento farmacológico , Expressão Gênica/efeitos dos fármacos , Miocárdio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Antagonistas Adrenérgicos beta/farmacologia , Adulto , Idoso , ATPases Transportadoras de Cálcio/efeitos dos fármacos , ATPases Transportadoras de Cálcio/genética , ATPases Transportadoras de Cálcio/metabolismo , Carbazóis/farmacologia , Carbazóis/uso terapêutico , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/fisiopatologia , Carvedilol , Feminino , Hemodinâmica , Humanos , Masculino , Metoprolol/farmacologia , Metoprolol/uso terapêutico , Pessoa de Meia-Idade , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Propanolaminas/farmacologia , Propanolaminas/uso terapêutico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Adrenérgicos beta/genética , Volume Sistólico/efeitos dos fármacos , Miosinas Ventriculares/efeitos dos fármacos , Miosinas Ventriculares/genética , Miosinas Ventriculares/metabolismo
18.
J Muscle Res Cell Motil ; 23(4): 335-9, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12630708

RESUMO

Decreased estrogen production is associated with changes in the skeletal, cardiovascular and muscular systems. At the level of skeletal muscles, it has been shown that a reduction in force production occurs at menopause but the underlying mechanisms are still unknown. The aim of the study was to investigate the effects of ovariectomy on myosin heavy chain (MyHC) composition. Additionally, we studied the effects of physical activity and the combined effects of physical activity and estrogen treatment on MyHC content in ovariectomised (OX) animals. Twenty-five rats were randomly assigned to five different groups: controls, runners, OX, ovariectomised runners and ovariectomised runners receiving estrogen. Exercise consisted of voluntary running for 5 weeks. Two muscles were analysed: m. extensor digitorum longus, EDL, (fast muscle) and m. soleus (slow muscle). MyHC content was analysed on 8% gel electrophoresis. The level of running activity is reduced in OX animals and estrogen administration is associated with the normalisation of the level of physical activity. Ovariectomy induces a shift from fast to slow MyHC isoforms in both the soleus and EDL. When OX animals are allowed to run, alterations in MyHC isoforms are still observed in the EDL but not in the soleus. When physical activity is combined with estrogen treatment no alterations are observed in both muscles. In conclusion, this study shows that ovariectomy induces alterations in the contractile properties of skeletal muscles and that physical activity in combination with estrogen treatment are associated with the maintenance of slow and fast muscle characteristics.


Assuntos
Estrogênios/deficiência , Fibras Musculares de Contração Rápida/metabolismo , Fibras Musculares de Contração Lenta/metabolismo , Debilidade Muscular/etiologia , Músculo Esquelético/metabolismo , Condicionamento Físico Animal , Pós-Menopausa/metabolismo , Animais , Estrogênios/farmacologia , Feminino , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Fibras Musculares de Contração Rápida/citologia , Fibras Musculares de Contração Rápida/efeitos dos fármacos , Fibras Musculares de Contração Lenta/citologia , Fibras Musculares de Contração Lenta/efeitos dos fármacos , Debilidade Muscular/metabolismo , Debilidade Muscular/fisiopatologia , Músculo Esquelético/citologia , Músculo Esquelético/efeitos dos fármacos , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Ovariectomia , Isoformas de Proteínas/efeitos dos fármacos , Isoformas de Proteínas/metabolismo , Ratos
19.
Circulation ; 104(6): 717-22, 2001 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-11489781

RESUMO

BACKGROUND: As shown previously in goats, clenbuterol increased the power of electrically conditioned skeletal muscle ventricles (SMVs) of clinically relevant size (150 mL), which were constructed around a mock system. They pumped against a pressure of 60 to 70 mm Hg immediately during surgery and up to several months after, finally at >1 L/min. SMVs without clenbuterol administration failed. Thus, we expected that clenbuterol-supported SMVs might become integrated into the circulation by a 1-step operation instead of the 2-step procedure required up to now. METHODS AND RESULTS: In adult Boer goats (n=5), latissimus dorsi muscle was wrapped around a polyurethane chamber of 150 mL that was connected to the descending aorta. This muscular flow-through pumping chamber containing a stabilizing inner layer (called a biomechanical heart [BMH]) was formed and immediately made to work against a systemic load with the support of clenbuterol (5x150 microg/wk). During surgery, the mean stroke volume of BMHs was 53.8+/-22.4 mL. One month after surgery, in peripheral arterial pressure, the mean diastolic (P(MD)) and minimal diastolic (P(min)) pressures of BMH-supported heart cycles differed significantly from unsupported ones (P(MD)=+2.9+/-1.1 mm Hg [P<0.04], P(min)=-2.4+/-0.9 mm Hg [P<0.04]). After BMH-supported heart contractions, the subsequent maximal rate of pressure generation, dP/dt(max), increased by 20.5+/-8.1% (P<0.02). One BMH, catheterized 132 days after surgery, shifted a volume of 34.8 mL per beat and 1.4 L/min with a latissimus dorsi muscle of 330 g. Depending on duration of training, the percentage of myosin heavy chain type 1 ranged between 31% and 100%. CONCLUSIONS: Under support of clenbuterol, BMHs of a clinically relevant size can be trained effectively in the systemic circulation after a 1-step operation and offer the prospect of a sufficient volume shift and probably unloading of the left ventricle.


Assuntos
Ventrículo de Músculo Esquelético , Animais , Fenômenos Biomecânicos , Pressão Sanguínea/efeitos dos fármacos , Clembuterol/farmacologia , Cabras , Masculino , Contração Muscular/efeitos dos fármacos , Músculo Esquelético/química , Músculo Esquelético/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Ventrículo de Músculo Esquelético/irrigação sanguínea , Ventrículo de Músculo Esquelético/fisiologia , Volume Sistólico/efeitos dos fármacos
20.
Biochem Biophys Res Commun ; 281(4): 924-8, 2001 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-11237749

RESUMO

The kinetics of actin-dependent MgATPase activity of skeletal muscle myosin subfragment 1 (S1) isoform containing the A1 essential light chain differ from those of the S1 isoform containing the A2 essential light chain. The differences are due to the presence of the extra N-terminal peptide comprising 42 amino acid residues in the A1 light chain. This peptide can interact with actin; heretofore, there have no been reports of the direct interaction between this peptide and the heavy chain of S1. Here, using the zero-length cross-linker 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) and S. aureus V8 protease, we show for the first time that the N-terminal part of the A1-light chain can interact with the 22-kDa fragment of the S1 heavy chain. No such interaction has been observed for the S1(A2) isoenzyme. Localization of residues which can possibly react with the cross-linker suggests that the interaction might involve the N-terminal residues of the A1 light chain and the converter region of the heavy chain.


Assuntos
Cadeias Pesadas de Miosina/metabolismo , Subfragmentos de Miosina/metabolismo , Actinas/farmacologia , Trifosfato de Adenosina/farmacologia , Sequência de Aminoácidos , Animais , Reagentes de Ligações Cruzadas , Eletroforese em Gel de Poliacrilamida , Etildimetilaminopropil Carbodi-Imida/química , Dados de Sequência Molecular , Músculo Esquelético/química , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/efeitos dos fármacos , Subfragmentos de Miosina/química , Subfragmentos de Miosina/efeitos dos fármacos , Ligação Proteica , Coelhos , Serina Endopeptidases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA