Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Semin Arthritis Rheum ; 56: 152051, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35780722

RESUMO

OBJECTIVES: The aim of this observational study was to compare clinical outcomes including glucocorticoid treatment and relapses between giant cell arteritis (GCA) patients with (axGCA) and without axillary artery involvement (non-axGCA). METHODS: Axillary artery ultrasound was performed in 101 GCA patients at multiple time points. Patients with signs of vasculitis of the axillary arteries at baseline were compared to patients without signs of axillary artery involvement. Cumulative GC doses and relapse rates were calculated as well as survival curves to compare the time until GC discontinuation and occurrence of the first clinical relapse. A linear mixed model was used to assess the effect of a clinical relapse on the intima media thickness (IMT) in axGCA patients. RESULTS: Sixty-seven patients were classified as axGCA, 34 as non-axGCA patients. Compared with non-axGCA, axGCA patients yielded a higher (albeit not significant) median time until GC discontinuation (42 months (95% CI: 33-84) vs 30 months (95% CI: 21-42), p=0.060) and median cumulative GC dose (6801mg (range 1748-34169) vs 5633mg (range: 2553-19967), p=0.051). Time until the first relapse (axGCA: 12 months (95% CI: 8-42) vs non-axGCA: 13.5 months (95% CI: 6-27), p=0522) and relapse rates (2 (range: 0-16) vs 1 (range: 0-13), p=0.67) were similar in both groups. Relapses resulted in an increase of the IMT by 0.18mm (95% CI: 0.07-0.30, p=0.003). CONCLUSION: Patients with axGCA have a trend towards longer treatment duration and higher GC requirements as compared to non-axGCA patients. A relapse leads to an increase of the IMT by 0.18mm.


Assuntos
Arterite de Células Gigantes , Artéria Axilar/diagnóstico por imagem , Espessura Intima-Media Carotídea , Arterite de Células Gigantes/diagnóstico por imagem , Arterite de Células Gigantes/tratamento farmacológico , Glucocorticoides/uso terapêutico , Humanos , Recidiva
2.
Eur J Appl Physiol ; 121(12): 3379-3387, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34477931

RESUMO

PURPOSE: Induction of IDO depends on the activation of AhR forming the AhR/IDO axis. Activated AhR can transcribe various target genes including cytotoxic and inhibiting receptors of NK cells. We investigated whether AhR and IDO levels as well as activating (NKG2D) and inhibiting (KIR2DL1) NK cell receptors are influenced by acute exercise and different chronic endurance exercise programs. METHODS: 21 adult breast and prostate cancer patients of the TOP study (NCT02883699) were randomized to intervention programs of 12 weeks of (1) endurance standard training or (2) endurance polarized training after a cardiopulmonary exercise test (CPET). Serum was collected pre-CPET, immediately post-CPET, 1 h post-CPET and after 12 weeks post-intervention. Flow cytometry analysis was performed on autologous serum incubated NK-92 cells for: AhR, IDO, KIR2DL1 and NKG2D. Differences were investigated using analysis-of-variance for acute and analysis-of-covariance for chronic effects. RESULTS: Acute exercise: IDO levels changed over time with a significant increase from post-CPET to 1 h post-CPET (p = 0.03). KIR2DL1 levels significantly decreased over time (p < 0.01). NKG2D levels remained constant (p = 0.31). Chronic exercise: for both IDO and NKG2D a significant group × time interaction, a significant time effect and a significant difference after 12 weeks of intervention were observed (IDO: all p < 0.01, NKG2D: all p > 0.05). CONCLUSION: Both acute and chronic endurance training may regulate NK cell function via the AhR/IDO axis. This is clinically relevant, as exercise emerges to be a key player in immune regulation.


Assuntos
Treino Aeróbico , Terapia por Exercício/métodos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Células Matadoras Naturais/metabolismo , Cinurenina/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Neoplasias da Mama/reabilitação , Células Cultivadas , Teste de Esforço , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias da Próstata/reabilitação , Triptofano Oxigenase/metabolismo
3.
Thromb Haemost ; 112(6): 1110-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25354936

RESUMO

Despite optimal therapy, the morbidity and mortality of patients presenting with an acute myocardial infarction (MI) remain significant, and the initial mechanistic trigger of myocardial "ischaemia/reperfusion (I/R) injury" remains greatly unexplained. Here we show that factors released from the damaged cardiac tissue itself, in particular extracellular RNA (eRNA) and tumour-necrosis-factor α (TNF-α), may dictate I/R injury. In an experimental in vivo mouse model of myocardial I/R as well as in the isolated I/R Langendorff-perfused rat heart, cardiomyocyte death was induced by eRNA and TNF-α. Moreover, TNF-α promoted further eRNA release especially under hypoxia, feeding a vicious cell damaging cycle during I/R with the massive production of oxygen radicals, mitochondrial obstruction, decrease in antioxidant enzymes and decline of cardiomyocyte functions. The administration of RNase1 significantly decreased myocardial infarction in both experimental models. This regimen allowed the reduction in cytokine release, normalisation of antioxidant enzymes as well as preservation of cardiac tissue. Thus, RNase1 administration provides a novel therapeutic regimen to interfere with the adverse eRNA-TNF-α interplay and significantly reduces or prevents the pathological outcome of ischaemic heart disease.


Assuntos
Comunicação Autócrina/efeitos dos fármacos , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/enzimologia , Miócitos Cardíacos/efeitos dos fármacos , RNA/metabolismo , Ribonucleases/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Antioxidantes/metabolismo , Hipóxia Celular , Citoproteção , Modelos Animais de Doenças , Camundongos , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/enzimologia , Mitocôndrias Cardíacas/patologia , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/imunologia , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/imunologia , Miocárdio/patologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/patologia , RNA/genética , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Fator de Necrose Tumoral alfa/imunologia
4.
Anaesthesist ; 63(2): 129-34, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24499961

RESUMO

BACKGROUND: Candida infections represent a relevant risk for patients in intensive care units resulting in increased mortality. Echinocandins have become the agents of choice for early and specific antifungal treatment in critically ill patients. Due to cardiac effects following echinocandin administration seen in intensive care unit (ICU) patients the in vitro effects of echinocandins and fluconazole on isolated cardiomyocytes of the rat were examined. AIM: The study was designed to investigate a possible impact of echinocandins and fluconazole in clinically relevant concentrations on the in vitro contractile responsiveness and shape of isolated rat cardiomyocytes. MATERIAL AND METHODS: Ventricular cardiomyocytes were isolated from Lewis rats. Cardiomyocytes were cultured in the presence of all licensed echinocandin preparations and fluconazole at concentrations of 0 (control), 0.1, 1, 3.3, 10, 33 and 100 µg/ml for 90 min. Cells were stimulated by biphasic electrical stimuli and contractile responsiveness was measured as shortening amplitude. Additionally, the ratio of rod-shaped to round cells was determined. RESULTS: Anidulafungin concentrations of 3.3 and 10 µg/ml caused a significant increase in contractile responsiveness, caspofungin showed a significant decrease at 10 µg/ml and micafungin concentrations of 3.3-33 µg/ml led to a significant increase in cell shortening. Measurement was not possible at 33 µg/ml for anidulafungin and caspofungin and at 100 µg/ml for all echinocandins due to a majority of round-shaped, non-contracting cardiomyocytes. Fluconazole showed no significant effect on cell shortening at all concentrations tested. For the three echinocandins the ratio of round-shaped, non-contracting versus rod-shaped normal contracting cardiomyocytes increased in a dose-dependent manner. CONCLUSIONS: Echinocandins impact the in vitro contractility of isolated cardiomyocytes of rats. This observation could be of great interest in the context of antifungal treatment.


Assuntos
Antifúngicos/farmacologia , Equinocandinas/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Animais , Antifúngicos/toxicidade , Separação Celular , Forma Celular , Equinocandinas/toxicidade , Estimulação Elétrica , Fluconazol/farmacologia , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Técnicas In Vitro , Contração Miocárdica/efeitos dos fármacos , Ratos , Ratos Endogâmicos Lew
5.
J Mol Cell Cardiol ; 53(1): 73-81, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22564366

RESUMO

AIMS: Since adenine nucleotide translocase 1 (ANT1) overexpression improved cardiac function in rats with activated renin-angiotensin system (RAS) and angiotensin II is known to enhance transforming growth factor ß (TGFß) signaling in cardiomyocytes, we assumed that ANT1 might modulate the classical TGFß/SMAD pathway. We therefore investigated whether the cardioprotective effect of ANT1 overexpression suppresses TGFß(1)-induced apoptosis, whether mitochondrial permeability transition pore (MPTP) regulation is involved, and SMAD signaling pathway is affected. METHODS AND RESULTS: Ventricular cardiomyocytes isolated from wild-type (WT) and ANT1 transgenic rats were treated with the apoptosis-inducing agent TGFß(1) (1 ng/ml). TGFß(1) treatment of WT cells enhanced the number of apoptotic cells by 31.8 ± 11.7% (p<0.01 vs. WT) measured by chromatin condensation. Apoptosis was blocked by 1µM cyclosporine A and by ANT1 overexpression. The protecting effect of ANT1 overexpression on TGFß(1)-induced apoptosis was verified by reduced caspase 3/7 activity and increased Bcl-2 expression. In addition, TGFß(1) decreased mitochondrial membrane potential as measured by JC-1 staining by 18.0 ± 3.7% in WT cardiomyocytes, but only by 7.2 ± 2.8% (p<0.05 vs. WT) in ANT1 cardiomyocytes. Cyclosporine A also attenuated the decline in mitochondrial membrane potential under TGFß(1) in WT cardiomyocytes. Determination of MPTP opening by Calcein assay in isolated cardiomyocytes and calcium retention assay in isolated mitochondria revealed a reduced open probability of MPTP after ANT1 overexpression. In addition to the effects of ANT1 on MPTP opening we investigated if ANT1 may interfere with the classical TGFß signaling pathway. Interestingly, ANT1-transgenic cardiomyocytes expressed less TGFß receptor II than WT cells. However, SMAD2 phosphorylation was already enhanced without TGFß(1) stimulation in these cells. Although no additional increase in SMAD2 phosphorylation was detectable after TGFß(1) treatment, SMAD signaling was still responsive to TGFß(1) indicated by an upregulation of SMAD7, a TGFß(1) target protein. CONCLUSION: Heart-specific overexpression of ANT1 leads to a reduced apoptotic response to TGFß(1) by preservation of the mitochondrial membrane potential, resistance to MPTP opening and altered TGFß signaling.


Assuntos
Apoptose/efeitos dos fármacos , Translocases Mitocondriais de ADP e ATP/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Animais , Apoptose/genética , Células Cultivadas , Expressão Gênica , Masculino , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Transgenes
6.
J Physiol Pharmacol ; 63(1): 17-22, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22460456

RESUMO

Mouse parathyroid hormone-related peptide (PTHrP) is a peptide hormone consisting of 139 amino acids. It is target of proteolysis resulting in circulation of N- and C-terminal peptides. C-terminal PTHrP peptides act in a PTH/PTHrP receptor-independent way with a minimal peptide sequence required to exert these effects covering amino acids 107-111 also known as osteostatin. Although effects of osteostatin on cardiac hypertrophy have been described in vitro, the in vivo relevance of these findings remained to be defined. The study was performed in two experimental series. In the first series, mice were randomly distributed into placebo or treatment group (each n=7) and osteostatin was administered via osmotic minipumps. In the second series, mice underwent a banding of the thoracic aorta to induce pressure overload and were again randomly distributed into placebo or treatment group (n=9 each). After 14 days, mice were anaesthetized and cardiac function, ECG, and cardiac hypertrophy were determined. Osteostatin increased the expression of ANF and reduced P-wave duration with little effects on cardiac performance in mice without pressure overload. In TAC banded mice, however, osteostatin significantly reduced TAC-induced loss of body weight, induced right ventricular hypertrophy, and reduced P-wave duration again. In osteostatin treated mice with pressure overload, the protein kinase C-dependent phosphorylation of connexin 43 was preserved. In summary, osteostatin attenuated pressure-overload-dependent loss of body weight without affecting left ventricular hypertrophy or left ventricular function but preserved atrial conduction. Osteostatin exerts moderate cardioprotective effects in mice under hemodynamic stress.


Assuntos
Aorta Torácica/fisiopatologia , Cardiomegalia/induzido quimicamente , Coração/efeitos dos fármacos , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/farmacologia , Função Ventricular Esquerda/fisiologia , Animais , Aorta Torácica/metabolismo , Fator Natriurético Atrial/metabolismo , Peso Corporal/efeitos dos fármacos , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia , Conexina 43/biossíntese , Conexina 43/genética , Conexina 43/metabolismo , Coração/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
7.
Minerva Cardioangiol ; 57(2): 203-17, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19274030

RESUMO

Cell based therapy has become a new and attractive option for the treatment of cardiac disease and heart failure. Although it has been demonstrated in vitro and in vivo that differentiation of non-differentiated cells (progenitor cells) into cardiomyocytes is able even in adult hearts the potential use of such transdifferentiation processes is limited by the small number of cells that home and engraft in the myocardium and complete the transdifferentiation process. Therefore, cell recruitment to the damaged heart is a major challenge to improve any cell based therapy. This process requires homing and engraftment of stem or progenitor cells. Major strategies to improve stem or progenitor cell homing are based on an improvement of stem or progenitor cell mobilization from the bone marrow. Strategies that have been shown to be successful are those that use granulocyte colony-stimulating factor (G-CSF). But although cell mobilization was indeed successful no major impact on hemodynamics was found. Alternatives are therefore needed and experimental studies use parathyroid hormone, statins, erythropoietin, and others in addition to or as an alternative to G-CSF. Although each of these procedures does have an impact on cell mobilization and homing none of these studies has provided a direct evidence that a major improvement on top of standard pharmacological therapy can be expected from such strategies. In conclusion, improvement of stem cell homing is a major challenge in the development of successful cell based therapies but not yet improved to a clinical relevant status. The underlying concepts of different strategies will be discussed here.


Assuntos
Mobilização de Células-Tronco Hematopoéticas/métodos , Infarto do Miocárdio/cirurgia , Transplante de Células-Tronco/métodos , Células-Tronco/efeitos dos fármacos , Animais , Eritropoetina/uso terapêutico , Fator Estimulador de Colônias de Granulócitos/farmacologia , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Metanálise como Assunto , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Hormônio Paratireóideo/uso terapêutico , Proteínas Recombinantes , Resultado do Tratamento
8.
J Physiol Pharmacol ; 60 Suppl 4: 5-13, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20083846

RESUMO

Although deficiency of the small leucine-rich proteoglycan decorin aggravates diabetic nephropathy in mice, the precise mechanisms of action are not fully understood. In the present study we used decorin-deficient mice (Dcn(-/-)) to further elucidate the molecular mechanisms involved in the protective action of decorin in diabetes. We discovered that streptozotocin-induced diabetes in Dcn(-/-) mice led to increased proteinuria associated with enhanced cyclin-dependent kinase inhibitor p27Kip1 in podocytes and tubular epithelial cells. Furthermore, lack of decorin increased the rate of apoptosis and caused overexpression of the IGF-IR in tubular epithelial cells of diabetic kidneys. In vitro experiments using human proximal renal epithelial cells showed that recombinant decorin was bound to the IGF-IR and protected against high glucose-mediated apoptosis. Furthermore, overexpression of TGFbeta1 and CTGF triggered by decorin deficiency resulted in enhanced accumulation of extracellular matrix in diabetic kidneys. Notably, diabetic Dcn(-/-) kidneys revealed marked upregulation of the proinflammatory proteoglycan biglycan and enhanced infiltration of mononuclear cells. Collectively, our results indicate that decorin is a protective agent during the development of diabetic nephropathy. Future therapeutic approaches that would either enhance the endogenous production of decorin or deliver recombinant decorin to the diseased kidney might improve the outcome of patients with diabetic nephropathy.


Assuntos
Apoptose/fisiologia , Diabetes Mellitus/metabolismo , Neuropatias Diabéticas/metabolismo , Neuropatias Diabéticas/patologia , Proteínas da Matriz Extracelular/deficiência , Monócitos/fisiologia , Proteoglicanas/deficiência , Animais , Northern Blotting , Western Blotting , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Decorina , Diabetes Mellitus/genética , Células Epiteliais/metabolismo , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/genética , Fibrose/metabolismo , Humanos , Rim/metabolismo , Camundongos , Camundongos Knockout , Infiltração de Neutrófilos/fisiologia , Podócitos/metabolismo , Polissacarídeos/metabolismo , Proteinúria/metabolismo , Proteoglicanas/genética , Receptor IGF Tipo 1/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
9.
Pharmacol Ther ; 119(3): 311-25, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18619489

RESUMO

Angiotensin II (Ang II) plays a major role in the progression of myocardial hypertrophy to heart failure. Inhibiting the angiotensin converting enzyme (ACE) or blockade of the corresponding Ang II receptors is used extensively in clinical practice, but there is scope for refinement of this mode of therapy. This review summarizes the current understanding of the direct effects of Ang II on cardiomyocytes and then focus particularly on interaction of components of the renin-angiotensin system with other hormones and cytokines. New findings described in approximately 400 papers identified in the PubMed database and published during the 2.5 years are discussed in the context of previous relevant literature. The cardiac action of Ang II is influenced by the activity of different isoforms of ACE leading to different amounts of Ang II by comparison with other angiotensinogen-derived peptides. The effect of Ang II is mediated by at least two different AT receptors that are differentially expressed in cardiomyocytes from neonatal, adult and failing hearts. The intracellular effects of Ang II are influenced by nitric oxide (NO)/cGMP-dependent cross talk and are mediated by the release of autocrine factors, such as transforming growth factor (TGF)-beta1 and interleukin (IL)-6. Besides interactions with cytokines, Ang II is involved in systemic networks including aldosterone, parathyroid hormone and adrenomedullin, which have their own effects on cardiomyocytes that modify, amplify or antagonize the primary effect of Ang II. Finally, hyperinsulemia and hyperglycaemia influence Ang II-dependent processes in diabetes and its cardiac sequelae.


Assuntos
Angiotensina II/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Angiotensina II/fisiologia , Animais , Apoptose/efeitos dos fármacos , Cardiomiopatia Hipertrófica/metabolismo , Cardiomiopatia Hipertrófica/patologia , Cardiomiopatia Hipertrófica/fisiopatologia , Humanos , Modelos Biológicos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de Angiotensina/metabolismo , Transdução de Sinais/fisiologia
10.
Eur Respir J ; 32(4): 881-91, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18550612

RESUMO

Alterations of the nitric oxide receptor, soluble guanylate cyclase (sGC) may contribute to the pathophysiology of pulmonary arterial hypertension (PAH). In the present study, the expression of sGC in explanted lung tissue of PAH patients was studied and the effects of the sGC stimulator BAY 63-2521 on enzyme activity, and haemodynamics and vascular remodelling were investigated in two independent animal models of PAH. Strong upregulation of sGC in pulmonary arterial vessels in the idiopathic PAH lungs compared with healthy donor lungs was demonstrated by immunohistochemistry. Upregulation of sGC was detected, similarly to humans, in the structurally remodelled smooth muscle layer in chronic hypoxic mouse lungs and lungs from monocrotaline (MCT)-injected rats. BAY 63-2521 is a novel, orally available compound that directly stimulates sGC and sensitises it to its physiological stimulator, nitric oxide. Chronic treatment of hypoxic mice and MCT-injected rats, with fully established PAH, with BAY 63-2521 (10 mg x kg(-1) x day(-1)) partially reversed the PAH, the right heart hypertrophy and the structural remodelling of the lung vasculature. Upregulation of soluble guanylate cyclase in pulmonary arterial smooth muscle cells was noted in human idiopathic pulmonary arterial hypertension lungs and lungs from animal models of pulmonary arterial hypertension. Stimulation of soluble guanylate cyclase reversed right heart hypertrophy and structural lung vascular remodelling. Soluble guanylate cyclase may thus offer a new target for therapeutic intervention in pulmonary arterial hypertension.


Assuntos
Regulação Enzimológica da Expressão Gênica , Guanilato Ciclase/biossíntese , Guanilato Ciclase/fisiologia , Hipertensão Pulmonar/enzimologia , Artéria Pulmonar/enzimologia , Receptores Citoplasmáticos e Nucleares/biossíntese , Receptores Citoplasmáticos e Nucleares/fisiologia , Animais , Modelos Animais de Doenças , Hemodinâmica , Humanos , Hipertrofia , Hipóxia , Imuno-Histoquímica/métodos , Camundongos , Monocrotalina/farmacologia , Pirimidinas/farmacologia , Ratos , Guanilil Ciclase Solúvel
11.
Pflugers Arch ; 457(2): 485-91, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18581135

RESUMO

alpha-Adrenergic stimulation results in a positive adaptation of cardiomyocytes to increased cardiac work load by induction of hypertrophy and enhanced contraction. However, sustained adrenergic stimulation causes progression to heart failure. Under simultaneous activation of alpha- and beta-adrenoceptors by the naturally occurring catecholamine noradrenaline, beta1-stimulation inhibits alpha-adrenergic-stimulated hypertrophy. If beta-adrenergic stimulation may also influence cardiomyocyte contraction is not known yet. We now demonstrate that exposure of cardiomyocytes to noradrenaline or isoprenaline for 24 h results in a reduced cell shortening at low beating frequencies (0.5 Hz). At high beating frequencies (2 Hz), cell shortening was normal. beta-adrenergic stimulation enhances SERCA2A expression at the messenger RNA and protein level. This induction of the Ca(2+) pump SERCA2A by the transcription factor NFAT is responsible for maintenance of normal cell contraction at high beating frequencies since inhibition of NFAT by decoy-oligonucleotides impairs SERCA2A expression and cell shortening after beta-adrenergic stimulation. In conclusion, although reduced cell shortening is found under low beating frequencies, we demonstrate preservation of cardiomyocyte contraction at 2 Hz after exposure to beta-adrenergic stimuli, which indicate that adrenergic stimulation a priori does not cause impaired heart function. The increase of SERCA expression indicates an even improved Ca(2+) handling of the cells.


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Cardiotônicos/farmacologia , Isoproterenol/farmacologia , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Norepinefrina/farmacologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/biossíntese , Animais , Cálcio/metabolismo , Estimulação Cardíaca Artificial , Células Cultivadas , Indução Enzimática , Frequência Cardíaca , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/enzimologia , Masculino , Miócitos Cardíacos/enzimologia , Fatores de Transcrição NFATC/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Fatores de Tempo
12.
Dtsch Med Wochenschr ; 133(12): 586-90, 2008 Mar.
Artigo em Alemão | MEDLINE | ID: mdl-18335390

RESUMO

Rapid interventional restoration of coronary blood flow is the most effective therapy to limit infarct size in today}s cardiology. The early phase of reperfusion represents, however, a window of therapeutic opportunities largely unused in the clinic. Experimentally it has been clearly shown that the modalities of reperfusion have a substantial impact on infarct size, since reperfusion itself can damage the myocardium (reperfusion injury). The major cause for acute injury of the cardiomyocytes in reperfusion is their hypercontracture. Cytosolic Ca (2+) overload and malfunction of cell organelles, i. e. sarcoplasmic reticulum and mitochondria, determine the pathophysiology of reperfusion injury. The underlying mechanisms can be influenced in the first minutes of reperfusion by activation of protective signalling pathways (reperfusion therapy). First clinical studies confirm the efficacy of acute reperfusion therapy.


Assuntos
Traumatismo por Reperfusão Miocárdica/fisiopatologia , Reperfusão Miocárdica/métodos , Cálcio/metabolismo , Circulação Coronária , Humanos , Mitocôndrias Cardíacas/metabolismo , Transdução de Sinais , Resultado do Tratamento
13.
Acta Physiol (Oxf) ; 191(4): 267-74, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17825088

RESUMO

AIM: Angiotensin-converting enzyme (ACE) inhibitors like ramiprilat bind to ACE expressed on the cell surface of endothelial cells and induce cell-specific signalling including the activation of activator protein (AP)-1. The present study addressed the question whether ramiprilat exerts a similar effect on adult ventricular cardiomyocytes, i.e. activates the AP-1 or modifies contractile performance. It was further aimed to decide whether such effects depend on bradykinin receptors or whether they are directly mediated via ACE. METHODS: Adult rat ventricular cardiomyocytes were isolated and cultured. mRNA expression of ACE was investigated by RT-PCR, AP-1 activation by gel mobility shift assays, and cardiac contractile performance by electrical pacing of isolated cells and analysis of cell shortening via a line-camera. RESULTS: Cardiomyocytes stably express ACE. Ramiprilat increased maximal contraction velocity and shortened the time-to-peak of contraction. In contrast to effects evoked by bradykinin, such effects caused by ramiprilat were not attenuated by HOE 140, a bradykinin-receptor antagonist. These effects were also not attenuated in the presence of l-nitro-arginine, used to mimic bradykinin-dependent signalling. In cardiomyocytes, bradykinin but not ramiprilat activated AP-1. Ramiprilat activates AP-1 in endothelial cells that are known to respond to ramiprilat in this way. CONCLUSION: Ramiprilat exerts direct, bradykinin-receptor independent effects on cardiomyocytes that improve cellular function without a corresponding effect on AP-1 activation or induction of AP-1 dependent effects. This newly described effect of ramiprilat may contribute to the protective effects seen by application of ACE inhibitors.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/administração & dosagem , Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Peptidil Dipeptidase A/metabolismo , Ramipril/análogos & derivados , Fator de Transcrição AP-1/metabolismo , Função Ventricular , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Ventrículos do Coração/citologia , Masculino , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Ramipril/administração & dosagem , Ratos , Ratos Wistar
14.
Cardiovasc Res ; 76(2): 311-22, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17658497

RESUMO

OBJECTIVE: Myocardial hypertrophy often develops in response to hypertension, and it is causal to and an independent predictor of heart failure. Several risk factors modify the progression of hypertrophy, the associated progressive impairment of myocardial function, and eventually the transition to overt congestive heart failure. The aim of the present study was to investigate the effects of smoking on the progression of pressure-dependent myocardial hypertrophy. METHODS: Spontaneously hypertensive rats (SHR) were used as a model for pressure-dependent hypertrophy. SHR were exposed to mainstream smoke from the Kentucky reference cigarette 2R4F (450 microg total particulate matter/l) or to fresh air (control), 5 days a week, twice for 1 h per day with a 30-minute fresh air exposure break for 30, 60, or 90 days. Endpoints for hypertrophy-associated changes were heart weight to body weight ratio, ventricular expression of hypertrophy-associated genes, ischemic tolerance, and inotropic responsiveness to isoprenaline in post-ischemic hearts. RESULTS: Smoke-exposed SHR showed a significant elevation in heart weight to body weight ratio, increased mRNA expression of atrial natriuretic factor (ANF), transforming growth factor (TGF)-beta(1), ornithine decarboxylase (ODC), and parathyroid hormone-related protein in both ventricles compared to controls. Hearts from smoke-exposed SHR showed a reduced recovery after 30 min global ischemia during the first 5 min of reperfusion and loss of inotropic stimulation after 30 min reperfusion. Smoke cessation was sufficient to reverse most of these alterations. WKY exposed to smoke did not develop similar changes. CONCLUSION: Our data indicate that several aspects of myocardial hypertrophy are accelerated by smoking.


Assuntos
Cardiomegalia/complicações , Insuficiência Cardíaca/etiologia , Hipertensão/complicações , Fumar/efeitos adversos , Animais , Progressão da Doença , Técnicas In Vitro , Masculino , Isquemia Miocárdica/fisiopatologia , Proteína Relacionada ao Hormônio Paratireóideo/genética , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptores Adrenérgicos beta/fisiologia , Fator de Crescimento Transformador beta1/fisiologia , Função Ventricular Esquerda
15.
Br J Pharmacol ; 151(6): 779-86, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17533419

RESUMO

BACKGROUND AND PURPOSE: Prostanoids have been shown to improve exercise tolerance, hemodynamics and quality of life in patients with pulmonary arterial hypertension (PAH). We investigated whether treprostinil exerts direct contractile effects on cardiomyocytes that may explain partly the beneficial effects of these drugs. EXPERIMENTAL APPROACH: Ventricular cardiomyocytes from adult rats were paced at a constant frequency of 0.5 to 2.0 Hz and cell shortening was monitored via a cell edge detection system. Twitch amplitudes, expressed as percent cell shortening of the diastolic cell length, and maximal contraction velocity, relaxation velocity, time to peak of contraction and time to reach 50% of relaxation were analyzed. KEY RESULTS: Treprostinil (0.15 - 15 ng ml(-1)) slightly increased contractile dynamics of cardiomyocytes at clinically relevant concentrations. However, the drug significantly improved cell shortening of cardiomyocytes in the presence of isoprenaline, a beta-adrenoceptor agonist. Treprostinil exerted this effect at all beating frequencies under investigation. Treprostinil mimicked this potentiating effect in a Langendorff preparation as well. The potentiating effect of treprostinil on isoprenaline-dependent cell shortening was no longer seen after phosphodiesterase inhibition. Long-term cultivation of cardiomyocytes with treprostinil did not modify load free cell shortening of these cells, but reduces the duration of contraction. CONCLUSIONS AND IMPLICATIONS: We conclude that the clinically used prostanoid treprostinil potentiates the positive inotropic effects of catecholamines in adult ventricular cardiomyocytes. This newly described effect may contribute to the beneficial clinical effects of prostanoids in patients with PAH.


Assuntos
Anti-Hipertensivos/farmacologia , Epoprostenol/análogos & derivados , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Agonistas Adrenérgicos beta/farmacologia , Animais , Anti-Hipertensivos/administração & dosagem , Catecolaminas/fisiologia , Tamanho Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Epoprostenol/administração & dosagem , Epoprostenol/farmacologia , Técnicas In Vitro , Isoproterenol/farmacologia , Masculino , Miócitos Cardíacos/metabolismo , Ratos , Ratos Wistar
16.
Basic Res Cardiol ; 100(4): 320-7, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15944810

RESUMO

Parathyroid hormone-related peptide (PTHrP) was found to improve contractile function of stunned myocardium in pigs. The peptide is released from coronary endothelial cells during ischemia and significantly improves post-ischemic recovery. The present study was aimed to decide whether such an induction of contractile responsiveness of the heart requires co-activation of adjacent cells or is a genuine phenomenon of cardiomyocytes. A second aim of this study was to decide whether such an improvement is linked to depressed cell function in general or oxidative inhibition. Isolated adult ventricular cardiomyocytes from rats were constantly paced at 0.5 Hz for 10 min. Cells were exposed to a brief oxidative inhibition by addition of 0.5 mmol/l potassium cyanide (KCN) in the presence of glucose. Under these conditions, cells stopped beating after 280 s on average. 30 s before they stopped to beat, cells had already developed a reduction in cell shortening, maximal relaxation and contraction velocity. In the co-presence of PTHrP (1-34) (100 nmol/l) cells continued to beat regular and did not develop reduced cell shortening, irrespectively of oxidative inhibition. In a second attempt, cells were exposed to the NO donor SNAP (100 micromol/l) or 8-bromocGMP (1 mmol/l). As expected both agents reduced cell shortening significantly. This reduction in cell shortening was attenuated in co-presence of PTHrP, too. Finally, we investigated the effect of PTHrP on cell shortening at different extracellular concentrations of calcium. Although, PTHrP increased intracellular calcium at 2 and 5 mmol/l extracellular calcium, respectively, it improved cell shortening only at 5 mmol/l extracellular calcium. Thus, the beneficial effect of PTHrP on cell shortening was independent from intracellular calcium but dependent on the steepness of the calcium gradient between intra- and extracellular calcium. In conclusion, our study shows that PTHrP is able to improve cell shortening rapidly and directly irrespectively of the reason for the reduced cell function. Improved electromechanical coupling rather than intracellular calcium handling seems to be the most important mechanism.


Assuntos
Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Animais , Cálcio/metabolismo , GMP Cíclico/fisiologia , Masculino , Miócitos Cardíacos/fisiologia , Óxido Nítrico/fisiologia , Oxirredução , Cianeto de Potássio/farmacologia , Ratos , Ratos Wistar
17.
Cardiovasc Res ; 66(1): 123-31, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15769455

RESUMO

OBJECTIVE: Reperfusion injury of the myocardium is characterised by development of cardiomyocyte hypercontracture. Previous studies have shown that cGMP-mediated stimuli protect against reperfusion injury, but the cellular mechanism is still unknown. METHODS: To simulate ischemia/reperfusion, adult rat cardiomyocytes were incubated anoxically (pH(o) 6.4) and then reoxygenated (pH(o) 7.4). Cytosolic calcium [Ca(2+)](i) (fura-2 ratio), pH(i) (BCECF ratio), cell length, and phospholamban phosphorylation were analysed. Under simulated ischemia cardiomyocytes develop [Ca(2+)](i) overload. When reoxygenated they rapidly undergo hypercontracture, triggered by oscillations of [Ca(2+)](i). We investigated whether cGMP-mediated stimuli can modulate [Ca(2+)](i) or pH(i) recovery and whether this contributes to their protective effect. Membrane-permeable cGMP analogues, 8-bromo-cGMP (1 mmol/L) or 8-pCPT-cGMP (10 micrommol/L), or a receptor-mediated activator of particulate guanylyl cyclase, urodilatin (1 micromol/L), were applied. RESULTS: The investigated stimuli protect against reoxygenation-induced hypercontracture (cell length as percent of end-ischemic length; control: 68+/-1.6; 8-bromo-cGMP: 88+/-1.5*; 8-pCPT-cGMP: 84+/-2.9*; urodilatin: 87+/-1.1*; n=24; *p<0.05). Recovery from [Ca(2+)](i) overload after 2 min reoxygenation [fura-2 ratio (a.u.); control: 1.43+/-0.15; 8-bromo-cGMP: 1.86+/-0.15*; 8-pCPT-cGMP: 1.92+/-0.19*; urodilatin: 1.93+/-0.24*; n=25; *p<0.05] was accelerated, and the frequency of [Ca(2+)](i) oscillations (min(-1)) was significantly reduced (control: 49+/-5.0 min(-1); 8-bromo-cGMP: 18+/-3.5* min(-1); 8-pCPT-cGMP: 18+/-4.5* min(-1); urodilatin: 16+/-4.1* min(-1); n=24; *p<0.05). cGMP-mediated stimuli increased sarcoplasmic Ca(2+) sequestration (caffeine-releasable Ca(2+) pool: 2-3 fold increase vs. control). Inhibition of sarcoplasmic Ca(2+)-ATPase (SERCA) by thapsigargin (150 nmol/L) or of protein kinase G with KT-5823 (1 micromol/L) abolished the effect of these stimuli on [Ca(2+)](i) recovery. The investigated stimuli significantly enhanced phospholamban phosphorylation. CONCLUSIONS: We conclude that cGMP-dependent signals activate SERCA via a protein kinase G-dependent phosphorylation of phospholamban. The increase in SERCA activity seems to reduce peak [Ca(2+)](i) and [Ca(2+)](i) oscillation during reoxygenation and to attenuate the excessive activation of the contractile machinery that otherwise leads to the development of hypercontracture.


Assuntos
Cálcio/metabolismo , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , GMP Cíclico/farmacologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Animais , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/metabolismo , Carbazóis/farmacologia , Tamanho Celular/efeitos dos fármacos , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Citosol/metabolismo , Concentração de Íons de Hidrogênio , Indóis/farmacologia , Masculino , Microscopia de Fluorescência , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Ratos Wistar , Retículo Sarcoplasmático/metabolismo , Tapsigargina/farmacologia
18.
Endocrinology ; 146(5): 2221-8, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15677763

RESUMO

Tuberoinfundibular peptide (TIP39) was initially identified as a neurotransmitter and agonist of the PTH2 receptor, which is expressed in the cardiovascular system. This study documents for the first time the cardiac expression and function of TIP39. Expression was analyzed via RT-PCR. Function was characterized on Langendorff-perfused rat hearts as left ventricular developed pressure (LVDP) and on isolated cells via a cell edge detection system. cGMP levels were detected with RIA. Tuberoinfundibular peptide (TIP39) mRNA was found to be constitutively expressed in coronary endothelium cells, isolated cardiomyocytes, ventricles, atria, and aorta. At first we investigated the vasodilatory properties of TIP39 (100 nM) in the presence of L-nitro-arginine (L-NA, 100 microM). Surprisingly, TIP39 had no vasodilatory effect but decreased LVDP by 35 +/- 7%. In the absence of L-NA, addition of TIP39 decreased LVDP by 8 +/- 2%. The PTH2 receptor antagonist Trp23-Tyr36-PTHrP (1-36, 100 nM) abolished this TIP39 effect in the presence of L-NA. The experiments with isolated cardiomyocytes provided similar results. TIP39 (10 nM) lowered the contraction amplitude by 6 +/- 3%. In the presence of L-NA (100 micromol/liter), TIP39 lowered the amplitude by 34 +/- 6%. cGMP concentration in cardiomyocytes was stimulated by TIP39 (10 nM) in the same range as by the nitric oxide (NO) donor SNAP (100 microM). In the presence of L-NA, this increase was abolished. These results suggest that an inhibition of endogenous NO production unmasks a profound negative inotropic effect of TIP39 that is mediated by an activation of the PTH2 receptor. The results obtained with isolated cardiomyocytes suggest that myocyte-derived NO, rather than vascular NO, is responsible for this effect. cGMP seems to be the downstream signal of produced NO.


Assuntos
Coração/fisiologia , Neuropeptídeos/fisiologia , Óxido Nítrico/biossíntese , Animais , Aorta/efeitos dos fármacos , Células Cultivadas , Vasos Coronários/efeitos dos fármacos , GMP Cíclico/análise , Depressão Química , Inibidores Enzimáticos/farmacologia , Expressão Gênica , Ventrículos do Coração/efeitos dos fármacos , Masculino , Contração Miocárdica/efeitos dos fármacos , Neuropeptídeos/genética , Neuropeptídeos/farmacologia , Óxido Nítrico/antagonistas & inibidores , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Nitroarginina/farmacologia , Pressão , Ratos , Ratos Wistar , Receptor Tipo 2 de Hormônio Paratireóideo/antagonistas & inibidores , Receptor Tipo 2 de Hormônio Paratireóideo/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vasodilatação/efeitos dos fármacos
19.
Am J Physiol Heart Circ Physiol ; 286(6): H2369-75, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-14764439

RESUMO

In some models of cardiac hypertrophy, activation of activator protein 1 (AP-1) correlates with growth. However, AP-1 is also activated by stimuli not involved in cardiac growth. This raises the following questions: does AP-1 plays a causal role for cardiomyocyte growth, and is this role model or stimulus dependent? We used a single model to address these questions, i.e., ventricular cardiomyocytes of adult rats, and two growth stimuli, i.e., alpha- and beta-adrenoceptor agonists [10 microM phenylephrine (PE) and 1 microM isoprenaline (Iso), respectively]. After 1 h of stimulation with PE, mRNA expression of c-Fos and c-Jun was upregulated to 185 +/- 32 and 132 +/- 13% of control. Fos and Jun proteins formed the AP-1 complex. PE stimulated DNA binding activity of AP-1 to 165 +/- 22% of control within 2 h and increased protein synthesis to 161 +/- 27% of control and cross-sectional area to 126 +/- 4% of control. Inhibition of AP-1 binding activity by cAMP response element (CRE) decoy oligonucleotides abolished both of these growth responses. Iso stimulated AP-1 binding activity to 203 +/- 19% of control within 2 h and stimulated protein synthesis to 145 +/- 17% of control. However, the growth effect of Iso was not abolished by CRE decoys: Iso increased protein synthesis to 158 +/- 17% of control in the presence of CRE. In conclusion, AP-1 is a causal mediator of the alpha-adrenergic, but not the beta-adrenergic, growth response of cardiomyocytes.


Assuntos
Cardiomegalia/fisiopatologia , Miócitos Cardíacos/fisiologia , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos beta/genética , Fator de Transcrição AP-1/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Células Cultivadas , Genes Precoces/fisiologia , Isoproterenol/farmacologia , Masculino , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Fenilefrina/farmacologia , RNA Mensageiro/análise , Ratos , Ratos Wistar
20.
Naunyn Schmiedebergs Arch Pharmacol ; 364(5): 444-53, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11692228

RESUMO

Adult rat ventricular cardiomyocytes contain alpha1A- and alpha1B-adrenoceptors (ARs, 20%:80%, assessed by [3H]prazosin binding). We studied which alpha1-AR subtype mediates noradrenaline (NA)-induced increase in rate of protein synthesis, and which signalling pathway is involved. NA (10-9-10-4 M) concentration-dependently increased inositol phosphate (IP) formation (pEC50-value=6.1+/-0.1, n=5) and protein synthesis (assessed as [3H]phenylalanine incorporation; pEC50-value=6.6+/-0.1, n=6). NA-induced IP-formation was partly inhibited by the alpha1B-AR antagonist chloroethylclonidine (CEC, 30 microM; 33+/-9% inhibition, n=5); following CEC-treatment the alpha1A-AR-selective 5-methyl-urapidil (5-MU) inhibited NA-induced IP-formation with a pKi-value of 9.2+/-0.2 (n=6); the alpha1D-AR-selective BMY 7378 was only a weak antagonist (pKi-value <7). NA-induced increase in protein synthesis was insensitive to CEC whereas 5-MU inhibited it with a pKi-value of 9.1+/-0.2 (n=6). NA (1 microM)-induced increase in protein synthesis was inhibited by the protein kinase C (PKC) inhibitor bisindolylmaleimide (IC50-value: 206 nM), the PI 3-kinase inhibitors wortmannin (IC50=3.4 nM) and LY 294002 (IC50=10 microM), and p70s6-kinase inhibitor rapamycin (IC50=123 pM) but not by the p38 MAP-kinase inhibitor SB 203580 (10 microM) or the MEK-inhibitor PD 98059 (25 microM). Moreover, 5-MU (30 nM) but not CEC inhibited NA-induced activation of p70s6-kinase. We conclude that, in adult rat cardiomyocytes, alpha1A- and alpha1B-AR mediate NA-induced IP-formation but only alpha1A-ARs mediate increase in protein synthesis. Alpha1A-AR-mediated increase in protein synthesis involves activation of a PKC, PI 3-kinase and p70s6-kinase but not of ERK- or p38 MAP-kinase.


Assuntos
Norepinefrina/farmacologia , Biossíntese de Proteínas , Receptores Adrenérgicos alfa 1/efeitos dos fármacos , Antagonistas de Receptores Adrenérgicos alfa 1 , Animais , Células Cultivadas , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Coração/efeitos dos fármacos , Fosfatos de Inositol/biossíntese , Masculino , Miocárdio/metabolismo , Ratos , Proteínas Quinases S6 Ribossômicas/antagonistas & inibidores , Sirolimo/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...