Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Métodos Terapêuticos e Terapias MTCI
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Ethnopharmacol ; 248: 112306, 2020 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-31626909

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Traditional Chinese medicine Leonurus japonicus Houtt. has a long history in the treatment of cardiovascular diseases. Stachydrine hydrochloride, the main bioactive ingredient extracted from Leonurus japonicus Houtt., has been shown to have cardioprotective effects. However, the underlying mechanisms of stachydrine hydrochloride haven't been comprehensively studied so far. AIM OF THE STUDY: The aim of this study was to investigate the protective role of stachydrine hydrochloride in heart failure and elucidate its possible mechanisms of action. MATERIALS AND METHODS: In vivo, transverse aorta constriction was carried out in C57BL/6J mice, and thereafter, 7.2 mg/kg telmisartan (a selective AT1R antagonist as positive control) and 12 mg/kg stachydrine hydrochloride was administered daily intragastrically for 4 weeks. Cardiac function was evaluated by assessing morphological changes as well as echocardiographic and haemodynamic parameters. In vitro, neonatal rat cardiomyocytes or adult mice cardiomyocytes were treated with stachydrine hydrochloride and challenged with phenylephrine (α-AR agonist). Ventricular myocytes were isolated from the hearts of C57BL/6J mice by Langendorff crossflow perfusion system. Intracellular calcium was measured by an ion imaging system. The length and movement of sarcomere were traced to evaluate the systolic and diastolic function of single myocardial cells. RESULTS: Stachydrine hydrochloride improved the cardiac function and calcium transient amplitudes, and inhibited the SR leakage and the amount of sparks in cardiac myocytes isolated from TAC mice. We also demonstrated that stachydrine hydrochloride could ameliorated phenylephrine-induced enhance in sarcomere contraction, calcium transients and calcium sparks. Moreover, our data shown that stachydrine hydrochloride blocked the hyper-phosphorylation of CaMKII, RyR2, PLN, and prevented the disassociation of FKBP12.6 from RyR2. CONCLUSION: Our results suggest that stachydrine hydrochloride exerts beneficial therapeutic effects against heart failure. These cardioprotective effects may be associated with the regulation of calcium handling by stachydrine hydrochloride through inhibiting the hyper-phosphorylation of CaMKII.


Assuntos
Aorta/fisiopatologia , Pressão Arterial , Sinalização do Cálcio/efeitos dos fármacos , Fármacos Cardiovasculares/farmacologia , Insuficiência Cardíaca/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Prolina/análogos & derivados , Função Ventricular Esquerda/efeitos dos fármacos , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Aorta/cirurgia , Proteínas de Ligação ao Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Fosforilação , Prolina/farmacologia , Ratos , Ratos Sprague-Dawley , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo , Telmisartan/farmacologia
2.
Physiol Rep ; 7(14): e14192, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31353833

RESUMO

Androgen therapy provides cardiovascular benefits for hypogonadism. However, myocardial hypertrophy, fibrosis, and infarction have been reported in testosterone or androgenic anabolic steroid abuse. Therefore, better understanding of the factors leading to adverse results of androgen abuse is needed. The aim of the present study was to examine the impact of high dose of androgen treatment on cardiac biology, and whether exposure duration modulates this response. Male rats were treated with 10 mg/kg testosterone, three times a week, for either 4 or 12 weeks; vehicle injections served as controls. Four weeks of testosterone treatment induced an increase in ventricular wall thickness, indicative of concentric hypertrophy, as well as increased ejection fraction; in contrast, both parameters were blunted following 12 weeks of high-dose testosterone treatment. Cardiac myocyte contractile parameters were assessed in isolated electrically stimulated myocytes (sarcomere and intracellular calcium dynamics), and in chemically permeabilized isolated myocardium (myofilament force development and tension-cost). High-dose testosterone treatment for 4 weeks was associated with increased myocyte contractile parameters, while 12 weeks treatment induced significant depression of these parameters, mirroring the cardiac pump function results. In conclusion, chronic administration of high-dose testosterone initially induces increased cardiac function. However, this initial beneficial impact is followed by significant depression of cardiac pump function, myocyte contractility, and cardiac myofilament function. Our results indicate that chronic high-testosterone usage is of limited use and may, instead, induce significant cardiac dysfunction.


Assuntos
Androgênios/farmacologia , Coração/efeitos dos fármacos , Contração Miocárdica , Testosterona/farmacologia , Androgênios/administração & dosagem , Androgênios/efeitos adversos , Animais , Cálcio/metabolismo , Células Cultivadas , Coração/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo , Sarcômeros/fisiologia , Testosterona/administração & dosagem , Testosterona/efeitos adversos
3.
J Cell Mol Med ; 22(10): 4830-4839, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30117672

RESUMO

MicroRNA-1 (miR-1) stands out as the most prominent microRNA (miRNA) in regulating cardiac function and has been perceived as a new potential therapeutic target. Lycium barbarum polysaccharides (LBPs) are major active constituents of the traditional Chinese medicine based on L. barbarum. The purpose of this study was to exploit the cardioprotective effect and molecular mechanism of LBPs underlying heart failure. We found that LBPs significantly reduced the expression of myocardial miR-1. LBPs improved the abnormal ECG and indexes of cardiac functions in P-V loop detection in transgenic (Tg) mice with miR-1 overexpression. LBPs recovered morphological changes in sarcomeric assembly, intercalated disc and gap junction. LBPs reversed the reductions of CaM and cMLCK, the proteins targeted by miR-1. Similar trends were also obtained in their downstream effectors including the phosphorylation of MLC2v and both total level and phosphorylation of CaMKII and cMyBP-C. Collectively, LBPs restored adverse structural remodelling and improved cardiac contractile dysfunction induced by overexpression of miR-1. One of the plausible mechanisms was that LBPs down-regulated miR-1 expression and consequently reversed miR-1-induced repression of target proteins relevant to myocardial contractibility. LBPs could serve as a new, at least a very useful adjunctive, candidate for prevention and therapy of heart failure.


Assuntos
Cardiotônicos/administração & dosagem , Medicamentos de Ervas Chinesas/administração & dosagem , MicroRNAs/genética , Contração Miocárdica/genética , Animais , Apoptose/efeitos dos fármacos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Cardiotônicos/química , Proteínas de Transporte/genética , Medicamentos de Ervas Chinesas/química , Regulação da Expressão Gênica/genética , Humanos , Medicina Tradicional Chinesa , Camundongos , Camundongos Transgênicos , Contração Miocárdica/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Sarcômeros/efeitos dos fármacos , Sarcômeros/genética , Sarcômeros/patologia
4.
Sci Rep ; 7: 44570, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28300163

RESUMO

Skeletal muscle tissue engineering holds great promise for pharmacological studies. Herein, we demonstrated an in vitro drug testing system using tissue-engineered skeletal muscle constructs. In response to epigenetic drugs, myotube differentiation of C2C12 myoblast cells was promoted in two-dimensional cell cultures, but the levels of contractile force generation of tissue-engineered skeletal muscle constructs prepared by three-dimensional cell cultures were not correlated with the levels of myotube differentiation in two-dimensional cell cultures. In contrast, sarcomere formation and contractile activity in two-dimensional cell cultures were highly correlated with contractile force generation of tissue-engineered skeletal muscle constructs. Among the epigenetic drugs tested, trichostatin A significantly improved contractile force generation of tissue-engineered skeletal muscle constructs. Follistatin expression was also enhanced by trichostatin A treatment, suggesting the importance of follistatin in sarcomere formation of muscular tissues. These observations indicate that contractility data are indispensable for in vitro drug screening.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Ácidos Hidroxâmicos/farmacologia , Engenharia Tecidual , Animais , Técnicas de Cultura de Células , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Folistatina/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Camundongos , Contração Muscular/efeitos dos fármacos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiologia , Mioblastos/efeitos dos fármacos , Sarcômeros/efeitos dos fármacos
5.
Biophys J ; 111(9): 2011-2023, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27806282

RESUMO

Actin-myosin cross-bridges use chemical energy from MgATP hydrolysis to generate force and shortening in striated muscle. Previous studies show that increases in sarcomere length can reduce thick-to-thin filament spacing in skinned muscle fibers, thereby increasing force production at longer sarcomere lengths. However, it is unclear how changes in sarcomere length and lattice spacing affect cross-bridge kinetics at fundamental steps of the cross-bridge cycle, such as the MgADP release rate. We hypothesize that decreased lattice spacing, achieved through increased sarcomere length or osmotic compression of the fiber via dextran T-500, could slow MgADP release rate and increase cross-bridge attachment duration. To test this, we measured cross-bridge cycling and MgADP release rates in skinned soleus fibers using stochastic length-perturbation analysis at 2.5 and 2.0 µm sarcomere lengths as pCa and [MgATP] varied. In the absence of dextran, the force-pCa relationship showed greater Ca2+ sensitivity for 2.5 vs. 2.0 µm sarcomere length fibers (pCa50 = 5.68 ± 0.01 vs. 5.60 ± 0.01). When fibers were compressed with 4% dextran, the length-dependent increase in Ca2+ sensitivity of force was attenuated, though the Ca2+ sensitivity of the force-pCa relationship at both sarcomere lengths was greater with osmotic compression via 4% dextran compared to no osmotic compression. Without dextran, the cross-bridge detachment rate slowed by ∼15% as sarcomere length increased, due to a slower MgADP release rate (11.2 ± 0.5 vs. 13.5 ± 0.7 s-1). In the presence of dextran, cross-bridge detachment was ∼20% slower at 2.5 vs. 2.0 µm sarcomere length due to a slower MgADP release rate (10.1 ± 0.6 vs. 12.9 ± 0.5 s-1). However, osmotic compression of fibers at either 2.5 or 2.0 µm sarcomere length produced only slight (and statistically insignificant) slowing in the rate of MgADP release. These data suggest that skeletal muscle exhibits sarcomere-length-dependent changes in cross-bridge kinetics and MgADP release that are separate from, or complementary to, changes in lattice spacing.


Assuntos
Difosfato de Adenosina/metabolismo , Contração Muscular/efeitos dos fármacos , Miosinas/metabolismo , Sarcômeros/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Fenômenos Biomecânicos/efeitos dos fármacos , Cálcio/farmacologia , Dextranos/farmacologia , Relação Dose-Resposta a Droga , Elasticidade/efeitos dos fármacos , Cinética , Locomoção/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Sarcômeros/efeitos dos fármacos , Sarcômeros/fisiologia , Viscosidade/efeitos dos fármacos
6.
Cardiovasc Pathol ; 24(4): 236-40, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25670255

RESUMO

BACKGROUND: Reactive oxygen species, such as superoxide, are being increasingly recognized as key components of a vast array of signaling pathways. Angiotensin II is a well-recognized stimulus for superoxide production through NADPH oxidase activation and opening of the mitochondrial ATP-sensitive potassium channels (mKATP). A role for this mechanism has been proposed to explain several physiological effects of the peptide. The aim of this study was to evaluate the involvement of this mechanism in the inotropic response to 100nmol/L angiotensin II. METHODS: Sarcomere shortening and intracellular pH (BCECF-epifluorescence technique) were evaluated in isolated cat ventricular myocytes placed in a perfusion chamber on the stage of an inverted microscope. Myocardial superoxide production was evaluated by the lucigenin quimioluminiscence method. RESULTS: Angiotensin II (100nmol/L) increased~70% sarcomere shortening, effect that was only partially prevented by NADPH oxidase inhibition, mKATP channel blockade or inhibition of the cardiac Na(+)/H(+) exchanger (NHE-1). Moreover, angiotensin II stimulates NHE-1 activity by a NADPH oxidase-dependent mechanism. Myocardial superoxide production was also increased by angiotensin II, and this action was completely prevented either by NADPH oxidase inhibition or mKATP channel blockade. CONCLUSIONS: The positive inotropic response to 100nmol/L angiotensin II is due to both ROS/NHE-1 dependent and independent pathways, this being a point of divergence with the signaling previously described to be triggered by lower concentrations of angiotensin II (i.e.: 1nmol/L).


Assuntos
Angiotensina II/farmacologia , Cardiotônicos/farmacologia , Ventrículos do Coração/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Superóxidos/metabolismo , Animais , Gatos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Ventrículos do Coração/metabolismo , Concentração de Íons de Hidrogênio , Miócitos Cardíacos/metabolismo , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Trocadores de Sódio-Hidrogênio/metabolismo
7.
Cell Rep ; 9(3): 810-21, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25437537

RESUMO

Diabetic cardiomyopathy is a complication of type 2 diabetes, with known contributions of lifestyle and genetics. We develop environmentally and genetically driven in vitro models of the condition using human-induced-pluripotent-stem-cell-derived cardiomyocytes. First, we mimic diabetic clinical chemistry to induce a phenotypic surrogate of diabetic cardiomyopathy, observing structural and functional disarray. Next, we consider genetic effects by deriving cardiomyocytes from two diabetic patients with variable disease progression. The cardiomyopathic phenotype is recapitulated in the patient-specific cells basally, with a severity dependent on their original clinical status. These models are incorporated into successive levels of a screening platform, identifying drugs that preserve cardiomyocyte phenotype in vitro during diabetic stress. In this work, we present a patient-specific induced pluripotent stem cell (iPSC) model of a complex metabolic condition, showing the power of this technique for discovery and testing of therapeutic strategies for a disease with ever-increasing clinical significance.


Assuntos
Cardiomiopatias Diabéticas/patologia , Avaliação Pré-Clínica de Medicamentos , Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Diferenciação Celular/efeitos dos fármacos , Humanos , Hipertrofia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Fenótipo , Sarcômeros/efeitos dos fármacos , Sarcômeros/patologia , Bibliotecas de Moléculas Pequenas/análise , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
8.
PLoS One ; 8(10): e78768, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24205315

RESUMO

Tamoxifen (Tam), a selective estrogen receptor modulator, is in wide clinical use for the treatment and prevention of breast cancer. High Tam doses have been used for treatment of gliomas and cancers with multiple drug resistance, but long QT Syndrome is a side effect. Tam is also used experimentally in mice for inducible gene knockout in numerous tissues, including heart; however, the potential direct effects of Tam on cardiac myocyte mechanical function are not known. The goal of this study was to determine the direct, acute effects of Tam, its active metabolite 4-hydroxytamoxifen (4OHT), and related drug raloxifene (Ral) on isolated rat cardiac myocyte mechanical function and calcium handling. Tam decreased contraction amplitude, slowed relaxation, and decreased Ca²âº transient amplitude. Effects were primarily observed at 5 and 10 µM Tam, which is relevant for high dose Tam treatment in cancer patients as well as Tam-mediated gene excision in mice. Myocytes treated with 4OHT responded similarly to Tam-treated cells with regard to both contractility and calcium handling, suggesting an estrogen-receptor independent mechanism is responsible for the effects. In contrast, Ral increased contraction and Ca²âº transient amplitudes. At 10 µM, all drugs had a time-dependent effect to abolish cellular contraction. In conclusion, Tam, 4OHT, and Ral adversely and differentially alter cardiac myocyte contractility and Ca²âº handling. These findings have important implications for understanding the Tam-induced cardiomyopathy in gene excision studies and may be important for understanding effects on cardiac performance in patients undergoing high-dose Tam therapy.


Assuntos
Cálcio/metabolismo , Contração Muscular/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Cloridrato de Raloxifeno/farmacologia , Tamoxifeno/análogos & derivados , Animais , Fenômenos Biomecânicos/efeitos dos fármacos , Feminino , Camundongos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Ratos , Ratos Sprague-Dawley , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo , Sarcômeros/fisiologia , Tamoxifeno/farmacologia
9.
Br J Pharmacol ; 169(5): 1178-88, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23578093

RESUMO

BACKGROUND AND PURPOSE: Doxorubicin is effective against breast cancer, but its major side effect is cardiotoxicity. The aim of this study was to determine whether the efficacy of doxorubicin on cancer cells could be increased in combination with PPARγ agonists or chrono-optimization by exploiting the diurnal cycle. EXPERIMENTAL APPROACH: We determined cell toxicity using MCF-7 cancer cells, neonatal rat cardiac myocytes and fibroblasts in this study. KEY RESULTS: Doxorubicin damages the contractile filaments of cardiac myocytes and affects cardiac fibroblasts by significantly inhibiting collagen production and proliferation at the level of the cell cycle. Cyclin D1 protein levels decreased significantly following doxorubicin treatment indicative of a G1/S arrest. PPARγ agonists with doxorubicin increased the toxicity to MCF-7 cancer cells without affecting cardiac cells. Rosiglitazone and ciglitazone both enhanced anti-cancer activity when combined with doxorubicin (e.g. 50% cell death for doxorubicin at 0.1 µM compared to 80% cell death when combined with rosiglitazone). Thus, the therapeutic dose of doxorubicin could be reduced by 20-fold through combination with the PPARγ agonists, thereby reducing adverse effects on the heart. The presence of melatonin also significantly increased doxorubicin toxicity, in cardiac fibroblasts (1 µM melatonin) but not in MCF-7 cells. CONCLUSIONS AND IMPLICATIONS: Our data show, for the first time, that circadian rhythms play an important role in doxorubicin toxicity in the myocardium; doxorubicin should be administered mid-morning, when circulating levels of melatonin are low, and in combination with rosiglitazone to increase therapeutic efficacy in cancer cells while reducing the toxic effects on the heart.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Doxorrubicina/administração & dosagem , PPAR gama/agonistas , Tiazolidinedionas/administração & dosagem , Animais , Animais Recém-Nascidos , Neoplasias da Mama/metabolismo , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Colágeno/metabolismo , Cronofarmacoterapia , Feminino , Fibroblastos/efeitos dos fármacos , Humanos , Ligantes , Células MCF-7 , Melatonina/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , PPAR gama/metabolismo , Ratos , Rosiglitazona , Sarcômeros/efeitos dos fármacos
10.
Micron ; 44: 298-302, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22917896

RESUMO

This work was undertaken to investigate the toxic activity of the post-synaptic neurotoxic fraction isolated from the venom of the Egyptian sand viper (Cerastes cerastes cerastes), and the ability of grape seed extract to antagonize this effect produced in sublethally intoxicated mice, with an emphasis on ultrastructural features. Light and transmission electron microscopy of diaphragms of intoxicated mice showed myonecrosis, myofiber hypercontraction, sarcomere disorganization, and mitochondrial damage. Alterations in motor neurons and axon terminals were also observed. The toxic activities of C. cerastes cerastes neurotoxin were inhibited by administrating grape seed extract, either before or after intoxication, showing that grape seed extract has protective and therapeutic potential to be used as antivenom.


Assuntos
Antivenenos/uso terapêutico , Diafragma/efeitos dos fármacos , Extrato de Sementes de Uva/uso terapêutico , Venenos de Víboras/toxicidade , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Antivenenos/farmacologia , Diafragma/metabolismo , Extrato de Sementes de Uva/farmacologia , Masculino , Camundongos , Microscopia Eletrônica de Transmissão , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/patologia , Miofibrilas/efeitos dos fármacos , Miofibrilas/patologia , Neurotoxinas , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/patologia , Sarcômeros/efeitos dos fármacos , Sarcômeros/patologia , Venenos de Víboras/farmacologia , Viperidae
11.
Am J Physiol Heart Circ Physiol ; 304(3): H465-72, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23203968

RESUMO

Alterations in intracellular Ca(2+) transients of cardiomyocytes in orchidectomized (ORX) rats could be a cause of cardiac dysfunction in the hypogonadal condition. To investigate the role of male sex hormones in intracellular Ca(2+) homeostasis during relaxation, Ca(2+)-handling activities by sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA) and the Na(+)/Ca(2+) exchanger (NCX) were evaluated in the ventricular muscle of 10-wk-old ORX rats with and without testosterone supplementation (2.5 mg/kg testosterone propionate, 2 times/wk). ORX induced a 50% decrease in contraction force accompanied by a prolonged time to achieve 50% relaxation (T(50)) in isolated intact ventricular trabeculae, which was partially corrected by testosterone administration. Maximum active tension was also suppressed in ORX rats without changes in myofilament Ca(2+) sensitivity and passive stiffness of the heart. Using a sarcoplasmic reticulum-enriched membrane preparation, the maximum thapsigargin-sensitive SERCA activity of the ORX rat was 27% lower with an increased Ca(2+) sensitivity, which was prevented by testosterone treatment. However, neither changes in SERCA content nor its modulating components, sarcolipin and heat shock protein 20, were detected in the ORX rat, but there was a significant decrease in the phosphorylated Thr(17) form of phospholamban. Despite a lower level of NCX protein in the heart of ORX rats, prolonged T(50) disappeared after an incubation with thapsigargin (10 µM), implying a lack of effect of male sex hormone deficiency on NCX function. These findings indicate that male sex hormones can regulate cardiac relaxation by acting mainly through SERCA. However, a detailed mechanism of SERCA modulation under male sex hormone deficiency status remains to be explored.


Assuntos
Contração Miocárdica/fisiologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/efeitos dos fármacos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/fisiologia , Trocador de Sódio e Cálcio/fisiologia , Testosterona/farmacologia , Animais , Western Blotting , Peso Corporal/efeitos dos fármacos , Coração/efeitos dos fármacos , Masculino , Contração Muscular/fisiologia , Proteínas Musculares/farmacologia , Músculo Esquelético/efeitos dos fármacos , Orquiectomia , Tamanho do Órgão/efeitos dos fármacos , Proteolipídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Sarcômeros/efeitos dos fármacos , Sarcômeros/ultraestrutura , Glândulas Seminais/efeitos dos fármacos , Testosterona/sangue , Testosterona/fisiologia , Malha Trabecular/fisiologia
12.
Cardiovasc Diabetol ; 11: 135, 2012 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-23116444

RESUMO

It has been reported previously that diabetic cardiomyopathy can be inhibited or reverted with chronic zinc supplementation. In the current study, we hypothesized that total cardiac calcium and zinc content is altered in early onset diabetes mellitus characterized in part as hyperglycemia (HG) and that exposure of zinc ion (Zn2+) to isolated cardiomyocytes would enhance contraction-relaxation function in HG more so than in nonHG controls. To better control for differential cardiac myosin isoform expression as occurs in rodents after ß-islet cell necrosis, hypothyroidism was induced in 16 rats resulting in 100% ß-myosin heavy chain expression in the heart. ß-Islet cell necrosis was induced in half of the rats by streptozocin administration. After 6 wks of HG, both HG and nonHG controls rats demonstrated similar myofilament performance measured as thin filament calcium sensitivity, native thin filament velocity in the myosin motility assay and contractile velocity and power. Extracellular Zn2+ reduced cardiomyocyte contractile function in both groups, but enhanced relaxation function significantly in the HG group compared to controls. Most notably, a reduction in diastolic sarcomere length with increasing pacing frequencies, i.e., incomplete relaxation, was more pronounced in the HG compared to controls, but was normalized with extracellular Zn2+ application. This is a novel finding implicating that the detrimental effect of HG on cardiomyocyte Ca2+ regulation can be amelioration by Zn2+. Among the many post-translational modifications examined, only phosphorylation of ryanodine receptor (RyR) at S-2808 was significantly higher in HG compared to nonHG. We did not find in our hypothyroid rats any differentiating effects of HG on myofibrillar protein phosphorylation, lysine acetylation, O-linked N-acetylglucosamine and advanced glycated end-products, which are often implicated as complicating factors in cardiac performance due to HG. Our results suggest that the relaxing effects of Zn2+ on cardiomyocyte function are more pronounced in the HG state due an insulin-dependent effect of enhancing removal of cytosolic Ca2+ via SERCA2a or NCX or by reducing Ca2+ influx via L-type channel or Ca2+ leak through the RyR. Investigations into the effects of Zn2+ on these mechanisms are now underway.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miosinas Ventriculares/metabolismo , Zinco/metabolismo , Animais , Glicemia/metabolismo , Cálcio/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/fisiopatologia , Diástole , Regulação da Expressão Gênica , Hipotireoidismo/metabolismo , Hipotireoidismo/fisiopatologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Masculino , Necrose , Isoformas de Proteínas , Processamento de Proteína Pós-Traducional , Ratos , Ratos Wistar , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Fatores de Tempo
13.
Clin Exp Pharmacol Physiol ; 39(1): 78-86, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21824171

RESUMO

1. Evidence is accumulating for a role for Ca²âº signalling in the differentiation and development of embryonic skeletal muscle. 2. Imaging of intact, normally developing transgenic zebrafish that express the protein component of the Ca²âº-sensitive complex aequorin, specifically in skeletal muscle, show that two distinct periods of spontaneous synchronised Ca²âº transients occur in the trunk: one at approximately 17.5-19.5 h post-fertilization (h.p.f.; termed signalling period SP1) and the other after approximately 23 h.p.f. (termed SP2). These periods of intense Ca²âº signalling activity are separated by a quiet period. 3. Higher-resolution confocal imaging of embryos loaded with the fluorescent Ca²âº reporter calcium green-1 dextran shows that the Ca²âº signals are generated almost exclusively in the slow muscle cells, the first muscle cells to differentiate, with distinct nuclear and cytoplasmic components. 4. Here, we show that coincidental with the SP1 Ca²âº signals, dystrophin becomes localized to the vertical myoseptae of the myotome. Introduction of a dmd morpholino (dmd-MO) resulted in no dystrophin being expressed in the vertical myoseptae, as well as a disruption of myotome morphology and sarcomere organization. In addition, the Ca²âº signalling signatures of dmd-MO-injected embryos or homozygous sapje mutant embryos were abnormal such that the frequency, amplitude and timing of the Ca²âº signals were altered compared with controls. 5. Our new data suggest that, in addition to a structural role, dystrophin may function in the regulation of [Ca²âº](i) during the early stages of slow muscle cell differentiation when the Ca²âº signals generated in these cells coincide with the first spontaneous contractions of the trunk.


Assuntos
Sinalização do Cálcio , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Desenvolvimento Muscular , Fibras Musculares de Contração Lenta/metabolismo , Fibras Musculares de Contração Lenta/patologia , Distrofias Musculares/metabolismo , Animais , Animais Geneticamente Modificados , Sinalização do Cálcio/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/patologia , Citoplasma/efeitos dos fármacos , Citoplasma/patologia , Desenvolvimento Embrionário/efeitos dos fármacos , Medições Luminescentes/métodos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microscopia Confocal , Microscopia de Fluorescência/métodos , Morfolinos/farmacologia , Contração Muscular/efeitos dos fármacos , Desenvolvimento Muscular/efeitos dos fármacos , Fibras Musculares de Contração Lenta/efeitos dos fármacos , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Distrofias Musculares/embriologia , Distrofias Musculares/patologia , Mutação , Especificidade de Órgãos , Transporte Proteico/efeitos dos fármacos , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo , Sarcômeros/patologia , Peixe-Zebra , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
14.
J Surg Res ; 165(1): 128-35, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20085844

RESUMO

BACKGROUND: Burn injury is frequently complicated by bacterial infection. Following burn injury, exposure to endotoxin produces a measurable decrease in cardiomyocyte sarcomere contractile function. Lipopolysaccharide-binding protein (LBP) is an acute phase protein that potentiates the recognition of lipopolysaccharide (LPS) by binding to the lipid A moiety of LPS. In this study, we sought to determine the effect of recombinant rat LBP (rLBP) on cardiomyocyte sarcomere function after burn or sham injury in the presence or absence of bacterial endotoxin. METHODS: Rats underwent a full-thickness 30% total body surface area scald or sham burn. At 24 h post-injury, cardiomyocytes were isolated, plated at 50,000 cells/well, and incubated with 50 µg/mL LPS and rLBP or chloramphenicol acetyltransferase (BVCat, an irrelevant control protein produced using the same expression system as rLBP) at concentrations by volume of 1%, 5%, 10%, and 30%. Subsets of cardiomyocytes were incubated with 5% rat serum or 30% rLBP and blocking experiments were conducted using an LBP-like synthetic peptide (LBPK95A). In vitro sarcomere function was measured using a variable rate video camera system with length detection software. RESULTS: Co-culture of burn and sham injury derived cardiomyocytes with high-dose rLBP in the presence of LPS resulted in a significant reduction to the functional impairment observed in peak sarcomere shortening following exposure to LPS alone. LBP-like peptide LBPK95A at a concentration of 20 µg/mL, in the presence of LPS, abolished the ability of 30% rLBP and 5% rat serum to restore peak sarcomere shortening of cardiomyocytes isolated following burn injury to levels of function exhibited in the absence of endotoxin exposure. CONCLUSIONS: In the setting of LPS challenge following burn injury, rLBP at high concentrations restores cardiomyocyte sarcomere contractile function in vitro. Rather than potentiating the recognition of LPS by the cellular LPS receptor complex, rLBP at high concentrations likely results in an inhibitory binding effect that minimizes the impact of endotoxin exposure on cardiomyocyte function following thermal injury.


Assuntos
Proteínas de Fase Aguda/farmacologia , Queimaduras/complicações , Proteínas de Transporte/farmacologia , Insuficiência Cardíaca/etiologia , Glicoproteínas de Membrana/farmacologia , Contração Miocárdica/efeitos dos fármacos , Animais , Apoptose , Sequência de Bases , Queimaduras/fisiopatologia , Relação Dose-Resposta a Droga , Marcação In Situ das Extremidades Cortadas , Lipopolissacarídeos/farmacologia , Masculino , Dados de Sequência Molecular , Miócitos Cardíacos/patologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Sarcômeros/efeitos dos fármacos , Sarcômeros/fisiologia
15.
Meat Sci ; 86(3): 699-705, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20619545

RESUMO

Beta agonists used as growth enhancers are known to affect the aging potential of beef muscle negatively. On the other hand, procedures like electrical stimulation could accelerate rigor and the aging process. In this study, 20 out of 40 young steers received no beta agonist (C), the remaining twenty steers received a beta agonist (zilpaterol hydrochloride) (Z) for the 30 days prior to slaughter followed by 4 days withdrawal. After slaughter carcasses were split, the left side electrically stimulated (ES) and the right side not stimulated (NES). Samples were aged for 3 or 14 days post mortem. Parameters included Warner Bratzler shear force (WBSF), myofibril filament length (MFL), sarcomere length and calpastatin and calpain enzyme activity. Zilpaterol resulted in increased (P<0.001) WBSF mainly due to an increased (P<0.001) calpastatin activity. ES improved tenderness (P<0.001) in general by early onset of rigor triggering the activity of calpains. ES also reduced the calpastatin activity (P<0.001), which partially countered the effect of high calpastatin activity on the aging potential of Z loins. ES can therefore be implemented to improve meat tenderness in zilpaterol supplemented steers, although steers without zilpaterol will still have an advantage in final tenderness.


Assuntos
Agonistas Adrenérgicos beta/efeitos adversos , Proteínas de Ligação ao Cálcio/metabolismo , Estimulação Elétrica , Tecnologia de Alimentos , Carne/análise , Músculo Esquelético/efeitos dos fármacos , Compostos de Trimetilsilil/efeitos adversos , Ração Animal , Animais , Calpaína/metabolismo , Bovinos , Masculino , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Miofibrilas/efeitos dos fármacos , Rigor Mortis , Sarcômeros/efeitos dos fármacos , Estresse Mecânico
16.
IDrugs ; 12(4): 243-51, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19350469

RESUMO

Cytokinetics Inc is developing CK-1827452, an agent that directly activates myosin, for use in the treatment of heart failure. The reaction of myosin cross-bridges from the thick myofilaments of cardiac sarcomeres with actin molecules from the thin myofilaments induces the muscle force and shortening that is responsible for systolic pressure development and ejection. Preclinical in vitro studies with isolated proteins investigated the mode of action of CK-1827452, demonstrating the ability of the drug to prolong the duration of time that myosin motors remain in a force-generating reaction with actin. Studies with isolated field-stimulated cardiac myocytes demonstrated the critical property of enhancing the extent of myocyte shortening, with no effect on the Ca2+ transient. Thus, CK-1827452 represents a novel myosin activator that acts independently of Ca2+, with an advantage compared with inotropic agents that elevate cAMP, enhance Ca2+ fluxes and have the potential to induce arrhythmias. Phase II clinical trials in patients with heart failure treated with intravenous and/or oral formulations of CK-1827452 support the hypothesis that the direct activation of myosin is a well-tolerated and viable approach to improving cardiac function. CK-1827452 represents a new and promising class of pure sarcomeric activators with a novel mechanism that could be an improvement over existing agents acting in part through this mechanism.


Assuntos
Cardiotônicos/uso terapêutico , Insuficiência Cardíaca/tratamento farmacológico , Miocárdio/metabolismo , Miosinas/metabolismo , Sarcômeros/efeitos dos fármacos , Doença Aguda , Cardiotônicos/administração & dosagem , Cardiotônicos/efeitos adversos , Cardiotônicos/farmacologia , Doença Crônica , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Avaliação Pré-Clínica de Medicamentos , Humanos , Sarcômeros/metabolismo , Resultado do Tratamento
17.
Circ Res ; 100(6): 864-73, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-17322173

RESUMO

Protein kinase D (PKD) is a serine/threonine kinase with emerging myocardial functions; in skinned adult rat ventricular myocytes (ARVMs), recombinant PKD catalytic domain phosphorylates cardiac troponin I at Ser22/Ser23 and reduces myofilament Ca(2+) sensitivity. We used adenoviral gene transfer to determine the effects of full-length PKD on protein phosphorylation, sarcomere shortening and [Ca(2+)](i) transients in intact ARVMs. In myocytes transduced to express wild-type PKD, the heterologously expressed enzyme was activated by endothelin 1 (ET1) (5 nmol/L), as reflected by PKD phosphorylation at Ser744/Ser748 (PKC phosphorylation sites) and Ser916 (autophosphorylation site). The ET1-induced increase in cellular PKD activity was accompanied by increased cardiac troponin I phosphorylation at Ser22/Ser23; this measured approximately 60% of that induced by isoproterenol (10 nmol/L), which activates cAMP-dependent protein kinase (PKA) but not PKD. Phosphorylation of other PKA targets, such as phospholamban at Ser16, phospholemman at Ser68 and cardiac myosin-binding protein C at Ser282, was unaltered. Furthermore, heterologous PKD expression had no effect on isoproterenol-induced phosphorylation of these proteins, or on isoproterenol-induced increases in sarcomere shortening and relaxation rate and [Ca(2+)](i) transient amplitude. In contrast, heterologous PKD expression suppressed the positive inotropic effect of ET1 seen in control cells, without altering ET1-induced increases in relaxation rate and [Ca(2+)](i) transient amplitude. Complementary experiments in "skinned" myocytes confirmed reduced myofilament Ca(2+) sensitivity by ET1-induced activation of heterologously expressed PKD. We conclude that increased myocardial PKD activity induces cardiac troponin I phosphorylation at Ser22/Ser23 and reduces myofilament Ca(2+) sensitivity, suggesting that altered PKD activity in disease may impact on contractile function.


Assuntos
Citoesqueleto de Actina/metabolismo , Cálcio/metabolismo , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Proteína Quinase C/fisiologia , Troponina I/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Animais , Cálcio/farmacologia , Células Cultivadas , Endotelina-1/farmacologia , Técnicas de Transferência de Genes , Genes Reporter , Proteínas de Fluorescência Verde/genética , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Isoproterenol/farmacologia , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/genética , Ratos , Sarcômeros/efeitos dos fármacos , Sarcômeros/fisiologia
18.
J Biol Chem ; 280(50): 41324-31, 2005 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-16236710

RESUMO

Cardiac troponin I is a phosphorylation target for endothelin-activated protein kinase C. Earlier work in cardiac myocytes expressing nonphosphorylatable slow skeletal troponin I provided evidence that protein kinase C-mediated cardiac troponin I phosphorylation accelerates relaxation. However, replacement with the slow skeletal isoform also alters the myofilament pH response and the Ca2+ transient, which could influence endothelin-mediated relaxation. Here, differences in the Ca2+ transient could not explain the divergent relaxation response to endothelin in myocytes expressing cardiac versus slow skeletal troponin I nor could activation of Na+/H+ exchange. Three separate clusters within cardiac troponin I are phosphorylated by protein kinase C, and we set out to determine the contribution of the Thr144 and Ser23/Ser24 clusters to the endothelin-mediated contractile response. Myocyte replacement with a cardiac troponin I containing a Thr144 substituted with the Pro residue found in slow skeletal troponin I resulted in prolonged relaxation in response to acute endothelin compared with control myocytes. Ser23/Ser24 also is a target for protein kinase C phosphorylation of purified cardiac troponin I, and although this cluster was not acutely phosphorylated in intact myocytes, significant phosphorylation developed within 1 h after adding endothelin. Replacement of Ser23/Ser24 with Ala indicated that this cluster contributes significantly to relaxation during more prolonged endothelin stimulation. Overall, results with these mutants provide evidence that Thr144 plays an important role in the acute acceleration of relaxation, whereas Ser23/Ser24 contributes to relaxation during more prolonged activation of protein kinase C by endothelin.


Assuntos
Endotelinas/química , Troponina I/química , Citoesqueleto de Actina/metabolismo , Adenoviridae/genética , Alanina/química , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Sítios de Ligação , Cálcio/metabolismo , Células Cultivadas , Análise por Conglomerados , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , DNA Complementar/metabolismo , Endotelinas/metabolismo , Concentração de Íons de Hidrogênio , Modelos Estatísticos , Células Musculares/citologia , Mutagênese , Contração Miocárdica , Miocárdio/citologia , Miocárdio/metabolismo , Fosforilação , Proteína Quinase C/metabolismo , Ratos , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo , Serina/química , Treonina/química , Fatores de Tempo , Troponina I/metabolismo
19.
Biofactors ; 25(1-4): 127-35, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16873937

RESUMO

Mechanisms underlying dilated cardiomyopathy (DCM) are poorly understood and effective therapy is still unavailable. The aim of this study was to examine the heart ultrastructure and dynamic of BIO T0-2 cardiomyopathic hamsters, an animal model of DCM, and to study in these animals, the effects of a co-formulation (HS12607) of propionyl-L-carnitine, coenzyme Q(10) and omega-3 fatty acids on cardiac mechanical parameters. Sarcomere length, Frank-Starling mechanism and force-frequency relations were studied on isolated ventricular papillary muscle from age-matched BIO F1B normal Syrian hamsters, BIO T0-2 control and BIO T0-2 HS12607-treated cardiomyopathic Syrian hamsters. At the optimum length to maximum active force, electron microscopy of left ventricular papillary muscle revealed that seven out of ten muscles studied showed shorter sarcomeres (1.20 +/- 0.29 microm), and the remaining three showed longer sarcomeres (2.80 +/- 0.13 microm), compared to those of normal hamsters (2.05 +/- 0.06 microm, n = 10). Severe alterations of the Frank-Starling mechanism, force-frequency relations and derivative parameters of contractile waves were also observed in vitro in the BIO T0-2 control hamsters. Long-term (8 weeks) treatment with HS12607 prevented alterations in sarcomere length in the BIO T0-2 cardiomyopathic hamsters; the Frank-Starling mechanism and force-frequency relations were also significantly (P < 0.05) improved in these hamsters. Therefore results of the present study strongly suggest the need for clinical studies on metabolic therapeutic intervention in the effort to stop the progression of dilated cardiomyopathy.


Assuntos
Cardiomiopatia Dilatada/tratamento farmacológico , Carnitina/análogos & derivados , Ácidos Graxos Ômega-3/uso terapêutico , Ubiquinona/análogos & derivados , Animais , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/fisiopatologia , Carnitina/uso terapêutico , Cricetinae , Modelos Animais de Doenças , Estimulação Elétrica , Masculino , Mesocricetus , Contração Miocárdica/efeitos dos fármacos , Músculos Papilares/efeitos dos fármacos , Sarcômeros/efeitos dos fármacos , Sarcômeros/ultraestrutura , Ubiquinona/uso terapêutico
20.
Biol Trace Elem Res ; 73(3): 193-200, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11049211

RESUMO

The beneficial effects of magnesium supplementation in pathological situations is well known, but the myocardial response to a nominal decrease in the level of magnesium has received relatively little attention. Hypomagnesemia can occur as chronic or acute manifestation of physiological changes, pathological conditions, or pharmacological interventions. Experimental interest was focused on the mechanical changes in adult rat heart myocytes following variation in extracellular Mg2+. Isolated cells were exposed to different levels of extracellular Mg2+ and the amplitude and rate of contraction were measured as a function of change in cell length using a video-based edge-detection system. Investigations have revealed that variation in the level of Mg2+ within physiological limits leads to mechanical changes. A decrease in the level of extracellular Mg2+ was accompanied by a significant increase in contractile amplitude and decrease in the velocities of contraction and relaxation. The contractile amplitude measured as percentage shortening were 3.08+/-0.19%, 4.62+/-0.19% and 6.9+/-0.40%, respectively, on exposure to 1.8, 0.8, and 0.48 mM Mg, and the corresponding velocities of contraction and relaxation normalized to amplitude were 0.54+/-0.02, 0.40+/-0.03, 0.31+/-0.03 and 0.47+/-0.02, 0.35+/-0.02, 0.24+/-0.02. The variations in contractile parameters associated with the change in the level of Mg were statistically significant (p < 0.01). Variation in the contractile properties associated with change in extracellular Mg2+ may be effected by alteration in Ca2+ transients.


Assuntos
Espaço Extracelular/fisiologia , Coração/fisiologia , Magnésio/fisiologia , Contração Miocárdica/fisiologia , Miocárdio/citologia , Animais , Frequência Cardíaca/efeitos dos fármacos , Técnicas In Vitro , Masculino , Ratos , Ratos Sprague-Dawley , Sarcômeros/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA