Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Purinergic Signal ; 13(1): 27-49, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27696085

RESUMO

Influences of adenosine 2A receptor (A2AR) activity on the cardiac transcriptome and genesis of endotoxemic myocarditis are unclear. We applied transcriptomic profiling (39 K Affymetrix arrays) to identify A2AR-sensitive molecules, revealed by receptor knockout (KO), in healthy and endotoxemic hearts. Baseline cardiac function was unaltered and only 37 A2AR-sensitive genes modified by A2AR KO (≥1.2-fold change, <5 % FDR); the five most induced are Mtr, Ppbp, Chac1, Ctsk and Cnpy2 and the five most repressed are Hp, Yipf4, Acta1, Cidec and Map3k2. Few canonical paths were impacted, with altered Gnb1, Prkar2b, Pde3b and Map3k2 (among others) implicating modified G protein/cAMP/PKA and cGMP/NOS signalling. Lipopolysaccharide (LPS; 20 mg/kg) challenge for 24 h modified >4100 transcripts in wild-type (WT) myocardium (≥1.5-fold change, FDR < 1 %); the most induced are Lcn2 (+590); Saa3 (+516); Serpina3n (+122); Cxcl9 (+101) and Cxcl1 (+89) and the most repressed are Car3 (-38); Adipoq (-17); Atgrl1/Aplnr (-14); H19 (-11) and Itga8 (-8). Canonical responses centred on inflammation, immunity, cell death and remodelling, with pronounced amplification of toll-like receptor (TLR) and underlying JAK-STAT, NFκB and MAPK pathways, and a 'cardio-depressant' profile encompassing suppressed ß-adrenergic, PKA and Ca2+ signalling, electromechanical and mitochondrial function (and major shifts in transcripts impacting function/injury including Lcn2, S100a8/S100a9, Icam1/Vcam and Nox2 induction, and Adipoq, Igf1 and Aplnr repression). Endotoxemic responses were selectively modified by A2AR KO, supporting inflammatory suppression via A2AR sensitive shifts in regulators of NFκB and JAK-STAT signalling (IκBζ, IκBα, STAT1, CDKN1a and RRAS2) without impacting the cardio-depressant gene profile. Data indicate A2ARs exert minor effects in un-stressed myocardium and selectively suppress NFκB and JAK-STAT signalling and cardiac injury without influencing cardiac depression in endotoxemia.


Assuntos
Endotoxemia/metabolismo , Miocárdio/metabolismo , Receptor A2A de Adenosina/metabolismo , Animais , Endotoxemia/genética , Perfilação da Expressão Gênica , Inflamação/genética , Inflamação/metabolismo , Janus Quinase 1/metabolismo , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Receptor A2A de Adenosina/genética , Fatores de Transcrição STAT/metabolismo , Transcriptoma
2.
Int J Cardiol ; 166(3): 672-80, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22192288

RESUMO

BACKGROUND: Cardiac injury and dysfunction are contributors to disease progression and mortality in sepsis. This study evaluated the cardiovascular role of intrinsic A2A adenosine receptor (A2AAR) activity during lipopolysaccharide (LPS)-induced inflammation. METHODS: We assessed the impact of 24 h of LPS challenge (20 mg/kg, IP) on cardiac injury, coronary function and inflammatory mediator levels in Wild-Type (WT) mice and mice lacking functional A2AARs (A2AAR KO). RESULTS: Cardiac injury was evident in LPS-treated WTs, with ~7-fold elevation in serum cardiac troponin I (cTnI), and significant ventricular and coronary dysfunction. Absence of A2AARs increased LPS-provoked cTnI release at 24 h by 3-fold without additional demise of contraction function. Importantly, A2AAR deletion per se emulated detrimental effects of LPS on coronary function, and LPS was without effect in coronary vessels lacking A2AARs. Effects of A2AAR KO were independent of major shifts in circulating C-reactive protein (CRP) and haptoglobin. Cytokine responses were largely insensitive to A2AAR deletion; substantial LPS-induced elevations (up to 100-fold) in IFN-γ and IL-10 were unaltered in A2AAR KO mice, as were levels of IL-4 and TNF-α. However, late elevations in IL-2 and IL-5 were differentially modulated by A2AAR KO (IL-2 reduced, IL-5 increased). Data demonstrate that in the context of LPS-triggered cardiac and coronary injury, A2AAR activity protects myocardial viability without modifying contractile dysfunction, and selectively modulates cytokine (IL-2, IL-5) release. A2AARs also appear to be targeted by LPS in the coronary vasculature. CONCLUSIONS: These experimental data suggest that preservation of A2AAR functionality might provide therapeutic benefit in human sepsis.


Assuntos
Cardiotônicos/metabolismo , Doença das Coronárias/metabolismo , Endotoxemia/metabolismo , Endotoxemia/prevenção & controle , Receptores A2 de Adenosina/deficiência , Animais , Doença das Coronárias/genética , Endotoxemia/genética , Feminino , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Knockout , Contração Miocárdica/fisiologia , Receptores A2 de Adenosina/genética
3.
J Pharmacol Exp Ther ; 335(1): 223-30, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20668052

RESUMO

The anthracycline doxorubicin (Dox) is an effective antitumor agent. However, its use is limited because of its toxicity in the heart. N-Benzyladriamycin-14-valerate (AD 198) is a modified anthracycline with antitumor efficacy similar to that of Dox, but with significantly less cardiotoxicity and potentially cardioprotective elements. In the present study, we investigated the possibility of in vivo protective effects of low-dose AD 198 against Dox-induced cardiomyopathy. To do this, rats were divided into four groups: vehicle, Dox (20 mg/kg; single injection day 1), AD 198 (0.3 mg/kg per injection; injections on days 1, 2, and 3), or a combination treatment of Dox + AD 198. Seventy-two hours after beginning treatment, hearts from the Dox group had decreased phosphorylation of AMP kinase and troponin I and reduced poly(ADP-ribose) polymerase, ß-tubulin, and serum albumin expression. Dox also increased the phosphorylation of phospholamban and expression of inducible nitric-oxide synthase in hearts. Each of these Dox-induced molecular changes was attenuated in the Dox + AD 198 group. In addition, excised hearts from rats treated with Dox had a 25% decrease in left ventricular developed pressure (LVDP) and a higher than normal increase in LVDP when perfused with a high extracellular Ca(2+) solution. The Dox-induced decrease in baseline LVDP and hyper-responsiveness to [Ca(2+)] was not observed in hearts from the Dox + AD 198 group. Thus Dox, with well established and efficient antitumor protocols, in combination with low levels of AD 198, to counter anthracycline cardiotoxicity, may be a promising next step in chemotherapy.


Assuntos
Antibióticos Antineoplásicos/antagonistas & inibidores , Antibióticos Antineoplásicos/toxicidade , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/prevenção & controle , Cardiotônicos/farmacologia , Doxorrubicina/antagonistas & inibidores , Doxorrubicina/toxicidade , Quinases Proteína-Quinases Ativadas por AMP , Animais , Western Blotting , Cálcio/farmacologia , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacologia , Eletroforese em Gel de Poliacrilamida , Indicadores e Reagentes , Masculino , Espectrometria de Massas , Miócitos Cardíacos/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/biossíntese , Fosforilação , Poli Adenosina Difosfato Ribose/farmacologia , Proteína Quinase C-épsilon/antagonistas & inibidores , Proteínas Quinases/metabolismo , Ratos , Ratos Wistar
4.
Am J Physiol Heart Circ Physiol ; 293(4): H2523-9, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17675570

RESUMO

Endogenous adenosine is an important ligand trigger for the cardioprotective effects of postconditioning (POC), yet it is unclear which adenosine receptor subtype is primarily responsible. To evaluate the role of A(2A) adenosine receptors in POC-induced protection, global ischemia-reperfusion was performed with and without POC in isolated wild-type (WT) and A(2A) adenosine receptor knockout (A(2A)KO) mouse hearts. Injury was measured in terms of postischemic functional recovery and release of cardiac troponin I (cTnI). Activation of protective signaling with POC was assessed by Akt and extracellular signal-regulated kinase (ERK) 1/2 phosphorylation. In WT hearts, POC improved recovery of postischemic developed pressure in early (81.6 +/- 6.4% of preischemic baseline vs. 37.5 +/- 5.6% for non-POC WT at 1 min) and late (62.2 +/- 4.2% of baseline vs. 45.5 +/- 5.3% for non-POC WT at 30 min) reperfusion, reduced cTnI release by 37%, and doubled the phosphorylation of both Akt and ERK1/2. These beneficial effects of POC were blocked by treatment with the selective A(2A) adenosine receptor antagonist ZM-241385 during reperfusion. Postischemic functional recovery, cTnI release, and phosphorylation of Akt and ERK1/2 were not different between non-POC WT and A(2A)KO hearts. In A(2A)KO hearts, POC did not improve functional recovery, reduce cTnI release, nor increase phosphorylation of Akt or ERK1/2. Thus the protective effects of POC are attenuated by both selective A(2A) receptor antagonism and targeted deletion of the gene encoding A(2A) adenosine receptors. These observations support the conclusion that endogenous activation of A(2A) adenosine receptors is an essential trigger leading to the protective effects of POC in isolated murine hearts.


Assuntos
Adenosina/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/metabolismo , Receptor A2A de Adenosina/metabolismo , Função Ventricular Esquerda , Antagonistas do Receptor A2 de Adenosina , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/enzimologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor A2A de Adenosina/deficiência , Receptor A2A de Adenosina/genética , Projetos de Pesquisa , Transdução de Sinais , Fatores de Tempo , Triazinas/farmacologia , Triazóis/farmacologia , Troponina I/metabolismo , Pressão Ventricular
5.
Am J Physiol Heart Circ Physiol ; 293(3): H1518-25, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17513494

RESUMO

Ischemia-reperfusion-induced Ca(2+) overload results in activation of calpain-1 in the heart. Calpain-dependent proteolysis contributes to myocardial dysfunction and cell death. Previously, preischemic treatment with low doses of H(2)O(2) was shown to improve postischemic function and reduce myocardial infarct size. Our aim was to determine the mechanism by which H(2)O(2) protects the heart. We hypothesized that H(2)O(2) causes the activation of p38 MAPK which initiates translocation of heat shock protein 25/27 (HSP25/27) to the myofilament Z disk. We further hypothesized that HSP25/27 shields structural proteins, particularly desmin, from calpain-induced proteolysis. To address this hypothesis, we first determined that an ischemia-reperfusion-induced decrease in desmin content could be blocked by H(2)O(2) pretreatment of hearts from rats. We next determined that ventricular myocytes that underwent Ca(2+) overload also demonstrated a calpain-dependent disruption of desmin that could be reduced by H(2)O(2)/p38 MAPK activation. Furthermore, myocytes acutely treated with H(2)O(2) exhibited a decrease in cleavage of desmin upon exposure to exogenous calpain-1 compared with myocytes not pretreated with H(2)O(2). The H(2)O(2)-induced attenuation of desmin degradation by calpain-1 was blocked by inhibition of p38 MAPK. In a final series of experiments, we demonstrated that cardiac myofilaments exposed to recombinant phosphorylated HSP27, but not nonphosphorylated HSP27, had a significant reduction in the calpain-induced degradation of desmin compared with non-HSP27-treated myofilaments. These findings are consistent with the hypothesis that H(2)O(2)-induced activation of p38 MAPK and subsequent HSP25/27 translocation attenuates desmin degradation brought about by calpain-1 activation in ischemia-reperfused hearts.


Assuntos
Calpaína/fisiologia , Desmina/metabolismo , Proteínas de Choque Térmico/metabolismo , Peróxido de Hidrogênio/farmacologia , Miócitos Cardíacos/metabolismo , Proteínas de Neoplasias/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Animais , Cálcio/farmacologia , Feminino , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Proteínas de Neoplasias/farmacologia , Ratos , Ratos Wistar , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Am J Physiol Heart Circ Physiol ; 292(2): H792-9, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17012362

RESUMO

The present study demonstrates that acute activation with either beta-adrenergic receptor agonists or H(2)O(2) treatment increases protein phosphatase 2a (PP2a) activity in ventricular myocytes. PP2a activation occurs concomitant with an increase in methylation of PP2a, changes in localization of a PP2a targeting subunit PP2aB56alpha, and a decrease in phosphorylation of PP2a substrates, such as troponin I (TnI) and ERK in ventricular myocytes. Okadaic acid, a well-established pharmacological inhibitor of PP2a, and the peptide Thr-Pro-Asp-Tyr-Phe-Leu (TPDYFL) were used to block PP2a methylation, localization, and phosphorylations. TPDYFL is a highly conserved sequence of the PP2a catalytic subunit COOH-terminus. Specifically, both okadaic acid and the peptide increased beta-adrenergic-cAMP-dependent phosphorylation of TnI and blocked the beta-adrenergic-cAMP-dependent translocation of PP2aB56alpha. TPDYFL, but not a scrambled version of this sequence, blocked H(2)O(2)-induced changes in PP2a methylation and TnI dephosphorylation. Okadaic acid produces similar inhibition of H(2)O(2) effects. Thus we propose that the novel peptide TPDYFL acts as an inhibitor of PP2a activity and may be a useful tool to increase our understanding of how PP2a is regulated and the role of PP2a in a variety of physiological and pathological processes. In addition, the present study is consistent with acute beta-adrenergic receptor activation and H(2)O(2) exposure, simultaneously activating kinases and PP2a to work on common substrates, such as TnI. We hypothesize that dual activation of opposing enzymes provides for a tighter regulation of substrate phosphorylations in ventricular myocytes.


Assuntos
Inibidores Enzimáticos/farmacologia , Miócitos Cardíacos/metabolismo , Oligopeptídeos/farmacologia , Fosfoproteínas Fosfatases/metabolismo , Troponina I/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Animais , Bucladesina/farmacologia , Células Cultivadas , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Peróxido de Hidrogênio/farmacologia , Metilação , Miócitos Cardíacos/efeitos dos fármacos , Ácido Okadáico/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 2 , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Wistar
7.
Am J Physiol Heart Circ Physiol ; 288(3): H1088-96, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15498825

RESUMO

We have proposed that pharmacological preconditioning, leading to PKC-epsilon activation, in hearts improves postischemic functional recovery through a decrease in actomyosin ATPase activity and subsequent ATP conservation. The purpose of the present study was to determine whether moderate PKC-independent decreases in actomyosin ATPase are sufficient to improve myocardial postischemic function. Rats were given propylthiouracil (PTU) for 8 days to induce a 25% increase in beta-myosin heavy chain with a 28% reduction in actomyosin ATPase activity. Recovery of postischemic left ventricular developed pressure (LVDP) was significantly higher in PTU-treated rat hearts subjected to 30 min of global ischemia than in control hearts: 57.9 +/- 6.2 vs. 32.6 +/- 5.1% of preischemic values. In addition, PTU-treated hearts exhibited a delayed onset of rigor contracture during ischemia and a higher global ATP content after ischemia. In the second part of our study, we demonstrated a lower maximal actomyosin ATPase and a higher global ATP content after ischemia in human troponin T (TnT) transgenic mouse hearts. In mouse hearts with and without a point mutation at F110I of human TnT, recovery of postischemic LVDP was 55.4 +/- 5.5 and 62.5 +/- 14.5% compared with 20.0 +/- 2.9% in nontransgenic mouse hearts after 35 min of global ischemia. These results are consistent with the hypothesis that moderate decreases in actomyosin ATPase activity result in net ATP conservation that is sufficient to improve postischemic contractile function.


Assuntos
Actomiosina/metabolismo , Metabolismo Energético/fisiologia , Isquemia Miocárdica/metabolismo , Miocárdio/metabolismo , Adenina/metabolismo , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/metabolismo , Creatina/metabolismo , Feminino , Humanos , Hipotireoidismo/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Cadeias Pesadas de Miosina/metabolismo , Proteína Quinase C/metabolismo , Proteína Quinase C-delta , Proteína Quinase C-épsilon , Ratos , Ratos Sprague-Dawley , Troponina T/genética , Troponina T/metabolismo
8.
Am J Physiol Heart Circ Physiol ; 285(6): H2578-86, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12881212

RESUMO

Evidence suggests that p38 mitogen-activated protein kinase (MAPK) activation influences cardiac function on an acute basis. The characterization and mechanisms by which this occurs were investigated in the present study. Adult rat ventricular myocytes treated with 1 mM arsenite for 30 min had a 16-fold increase in p38 MAPK phosphorylation that was attenuated by SB-203580 (a p38 MAPK inhibitor). Extracellular signal-regulated protein kinase (ERK) and c-Jun NH2-terminal kinase (JNK) were also minimally activated, but this activation was not sensitive to SB-203580. In addition, arsenite caused a p38 MAPK-independent translocation/activation of protein phosphatase 2a (PP2a) and decrease in phosphorylation of myosin light chain 2 (LC2). Arsenite-p38 MAPK activation led to translocation of heat shock protein 27 but not alpha B-crystallin to the myofilaments. Using isolated cardiomyocytes, we determined that arsenite reduces isometric tension without a change in Ca2+ sensitivity of tension via p38 MAPK and lowers myofibrillar actomyosin Mg2+-ATPase activity in a p38 MAPK-independent manner. Thus arsenite induces a p38 MAPK-independent change in PP2a and LC2 that may account for the arsenite-dependent decrease in ATPase and a p38 MAPK-dependent modification of the myofilaments that decreases myocardial force development.


Assuntos
Citoesqueleto de Actina/fisiologia , Proteínas de Choque Térmico , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Contração Miocárdica/fisiologia , Miócitos Cardíacos/enzimologia , Animais , Arsenitos/farmacologia , Miosinas Cardíacas/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Proteínas de Choque Térmico HSP27 , Ventrículos do Coração/citologia , Miócitos Cardíacos/efeitos dos fármacos , Cadeias Leves de Miosina/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Proteína Fosfatase 2 , Ratos , Cadeia B de alfa-Cristalina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
9.
Am J Physiol Heart Circ Physiol ; 285(3): H1220-8, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12763745

RESUMO

Activation of myocardial kappa-opioid receptor-protein kinase C (PKC) pathways may improve postischemic contractile function through a myofilament reduction in ATP utilization. To test this, we first examined the effects of PKC inhibitors on kappa-opioid receptor-dependent cardioprotection. The kappa-opioid receptor agonist U50,488H (U50) increased postischemic left ventricular developed pressure and reduced postischemic end-diastolic pressure compared with controls. PKC inhibitors abolished the cardioprotective effects of U50. To determine whether kappa-opioid-PKC-dependent decreases in Ca2+-dependent actomyosin Mg2+-ATPase could account for cardioprotection, we subjected hearts to three separate actomyosin ATPase-lowering protocols. We observed that moderate decreases in myofibrillar ATPase were equally cardioprotective as kappa-opioid receptor stimulation. Immunoblot analysis and confocal microscopy revealed a kappa-opioid-induced increase in myofilament-associated PKC-epsilon, and myofibrillar Ca2+-independent PKC activity was increased after kappa-opioid stimulation. This PKC-myofilament association led to an increase in troponin I and C-protein phosphorylation. Thus we propose PKC-epsilon activation and translocation to the myofilaments causes a decrease in actomyosin ATPase, which contributes to the kappa-opioid receptor-dependent cardioprotective mechanism.


Assuntos
(trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Citoesqueleto de Actina/enzimologia , Adenosina Trifosfatases/metabolismo , Analgésicos não Narcóticos/farmacologia , Isquemia Miocárdica/metabolismo , Proteína Quinase C/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cardiotônicos/farmacologia , Feminino , Precondicionamento Isquêmico Miocárdico , Isoenzimas/metabolismo , Isquemia Miocárdica/tratamento farmacológico , Fosforilação/efeitos dos fármacos , Proteína Quinase C-épsilon , Ratos , Ratos Wistar , Receptores Opioides kappa/metabolismo , Pressão Ventricular/efeitos dos fármacos
10.
Am J Physiol Heart Circ Physiol ; 285(1): H90-6, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12637352

RESUMO

Conflicting reports exist regarding the influence of beta-adrenergic stimulation on the maximum velocity of shortening (Vmax) in ventricular myocytes. This may be due to an unrecognized effect of maturation. In the present study, the effects of beta-adrenergic receptor stimulation on myocytes from hearts of juvenile nonbred and young adult retired breeder female rats were compared. Ventricular myocytes from young adults had a beta-adrenergic-dependent increase in Vmax and Ca2+-dependent actomyosin ATPase that was not observed in myocytes from juveniles. Myocytes from young adults had both an increase in beta-myosin heavy chain (MHC) and higher basal serine/threonine phosphatase activity compared with juvenile rats. Additional studies established moderate increases in beta-MHC induced by hypothyroidism do not confer myocardial beta-adrenergic responsiveness, whereas inhibition of the higher phosphatase activity in myocytes from young adults blocks the age-dependent, beta-adrenergic-induced increase in cross-bridge cycling rates. We propose that the higher phosphatase activity of myocytes from young adults compared with juveniles allows for a greater functional response of the myocardium to beta-adrenergic stimulation.


Assuntos
Envelhecimento/fisiologia , Células Musculares/enzimologia , Monoéster Fosfórico Hidrolases/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Western Blotting , Bucladesina/farmacologia , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/metabolismo , Inibidores Enzimáticos , Feminino , Ventrículos do Coração/citologia , Ventrículos do Coração/enzimologia , Hipotireoidismo/enzimologia , Técnicas In Vitro , Cinética , Contração Miocárdica/efeitos dos fármacos , Miocárdio/citologia , Miocárdio/enzimologia , Miofibrilas/enzimologia , Cadeias Pesadas de Miosina/metabolismo , Ácido Okadáico/farmacologia , Fosfatos/metabolismo , Radioisótopos de Fósforo , Fosforilação , Ratos
11.
Am J Physiol Heart Circ Physiol ; 285(1): H97-103, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12649078

RESUMO

Adenosine A1 receptor activation causes protein phosphatase 2a (PP2a) activation in ventricular myocytes. This attenuates beta-adrenergic functional effects in the heart (Liu Q and Hofmann PA. Am J Physiol Heart Circ Physiol 283: H1314-H1321, 2002). The purpose of the present study was to identify the signaling pathway involved in the translocation/activation of PP2a by adenosine A1 receptors in ventricular myocytes. We found that N6-cyclopentyladenosine (CPA; an adenosine A1 receptor agonist)-induced PP2a translocation was blocked by p38 MAPK inhibition but not by JNK inhibition. CPA increased phosphorylation of p38 MAPK, and this effect was abolished by pertussis toxin and inhibitors of the cGMP pathway. Moreover, CPA-induced PP2a translocation was blocked by inhibition of the cGMP pathway. Guanylyl cyclase activation mimicked the effects of CPA and caused p38 MAPK phosphorylation and PP2a translocation. Finally, CPA-induced dephosphorylations of troponin I and phospholamban were blocked by pertussis toxin and attenuated by p38 MAPK inhibition. These results suggest that adenosine A1 receptor-mediated PP2a activation uses a pertussis toxin-sensitive Gi protein-guanylyl cyclase-p38 MAPK pathway. This proposed, novel pathway may play a role in acute modulation of cardiac function.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células Musculares/metabolismo , Miocárdio/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Receptores Purinérgicos P1/metabolismo , Animais , Bucladesina/farmacologia , Proteínas de Ligação ao Cálcio/metabolismo , Ativação Enzimática/efeitos dos fármacos , Feminino , Guanilato Ciclase/metabolismo , Técnicas In Vitro , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Miocárdio/citologia , Proteína Fosfatase 2 , Antagonistas de Receptores Purinérgicos P1 , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Translocação Genética/genética , Troponina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
12.
Am J Physiol Heart Circ Physiol ; 283(4): H1314-21, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12234781

RESUMO

The ability of adenosine A(1) receptors to activate type 2a protein phosphatase (PP2a) and account for antiadrenergic effects was investigated in rat myocardial preparations. We observed that the adenosine A(1) receptor agonist N(6)-cyclopentyladenosine (CPA) significantly reduces the isoproterenol-induced increase in left ventricular developed pressure of isolated heats, and this effect is blocked by pretreatment of hearts with the PP2a inhibitor cantharidin. CPA alone or given in conjunction with isoproterenol stimulation decreases phosphorylation of phospholamban and troponin I in ventricular myocytes. These dephosphorylations are blocked by an adenosine A(1) receptor antagonist and by PP2a inhibition with okadaic acid. Adenosine A(1) receptor activation was also shown to increase carboxymethylation of the PP2a catalytic subunit (PP2a-C) and cause translocation of PP2a-C to the particulate fraction in ventricular myocytes. These results support the hypothesis that adenosine A(1) receptor activation leads to methylation of PP2a-C and subsequent translocation of the PP2a holoenzyme. Increases in localized PP2a activity lead to dephosphorylation of key cardiac proteins responsible for the positive inotropic effects of beta-adrenergic stimulation.


Assuntos
Adenosina/análogos & derivados , Miocárdio/enzimologia , Fosfoproteínas Fosfatases/metabolismo , Receptores Purinérgicos P1/metabolismo , Adenosina/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Cantaridina/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Técnicas In Vitro , Isoproterenol/farmacologia , Metilação , Fibras Musculares Esqueléticas/enzimologia , Miocárdio/citologia , Ácido Okadáico/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosforilação , Proteína Fosfatase 2 , Ratos , Ratos Wistar , Troponina I/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA