Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 32(36): 4191-202, 2013 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-23318436

RESUMO

Thalidomide and its analogues (lenalidomide and pomalidomide) are small molecule glutamic acid derivatives of the immunomodulatory drug (IMiD) class. In addition to the immuno-adjuvant and anti-inflammatory properties that define an IMiD, the thalidomide analogues demonstrate an overlapping and diverse range of biological activities, including anti-angiogenic, teratogenic and epigenetic effects. Importantly, the IMiDs possess anti-cancer activity with selectivity for molecularly defined subgroups of hematological malignancies, specifically mature B-cell neoplasms and myelodysplasia with deletion of chromosome 5q. Emerging insight into the pathophysiological drivers of these IMiD-responsive disease states can now be synthesized using previously disclosed IMiD activities and recently discovered thalidomide targets to build unifying models of IMiD mechanism of action. Attention to mechanisms of IMiD-induced clinical toxicities, in particular the recently identified association of lenalidomide with second primary malignancies, provides an additional tool for determination of drug mechanism. This review seeks to define the molecular IMiD targets and biological outputs that underpin their anti-neoplastic activity. It is anticipated that elucidation of important IMiD targets will allow the rational development of new-generation therapeutics with the potential to separate thalidomide-analogue efficacy from clinical toxicity.


Assuntos
Antineoplásicos/uso terapêutico , Fatores Imunológicos/uso terapêutico , Neoplasias/tratamento farmacológico , Talidomida/uso terapêutico , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/química , Deleção Cromossômica , Cromossomos Humanos Par 5 , Epigênese Genética/efeitos dos fármacos , Humanos , Fatores Imunológicos/efeitos adversos , Fatores Imunológicos/química , Imunomodulação/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/imunologia , Neoplasias/genética , Neoplasias/imunologia , Talidomida/efeitos adversos , Talidomida/análogos & derivados , Talidomida/química
2.
Oncogene ; 31(15): 1949-62, 2012 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-21909130

RESUMO

The phosphatidylinositol 3-kinase (PI3K)/AKT and RAS oncogenic signalling modules are frequently mutated in sporadic human cancer. Although each of these pathways has been shown to play critical roles in driving tumour growth and proliferation, their activation in normal human cells can also promote cell senescence. Although the mechanisms mediating RAS-induced senescence have been well characterised, those controlling PI3K/AKT-induced senescence are poorly understood. Here we show that PI3K/AKT pathway activation in response to phosphatase and tensin homolog (PTEN) knockdown, mutant PI3K, catalytic, α polypeptide (PIK3CA) or activated AKT expression, promotes accumulation of p53 and p21, increases cell size and induces senescence-associated ß-galactosidase activity. We demonstrate that AKT-induced senescence is p53-dependent and is characterised by mTORC1-dependent regulation of p53 translation and stabilisation of p53 protein following nucleolar localisation and inactivation of MDM2. The underlying mechanisms of RAS and AKT-induced senescence appear to be distinct, demonstrating that different mediators of senescence may be deregulated during transformation by specific oncogenes. Unlike RAS, AKT promotes rapid proliferative arrest in the absence of a hyperproliferative phase or DNA damage, indicating that inactivation of the senescence response is critical at the early stages of PI3K/AKT-driven tumourigenesis. Furthermore, our data imply that chronic activation of AKT signalling provides selective pressure for the loss of p53 function, consistent with observations that PTEN or PIK3CA mutations are significantly associated with p53 mutation in a number of human tumour types. Importantly, the demonstration that mTORC1 is an essential mediator of AKT-induced senescence raises the possibility that targeting mTORC1 in tumours with activated PI3K/AKT signalling may exert unexpected detrimental effects due to inactivation of a senescence brake on potential cancer-initiating cells.


Assuntos
Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Transformação Celular Neoplásica/genética , Células Cultivadas , Senescência Celular/genética , Dano ao DNA , Fibroblastos , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos , PTEN Fosfo-Hidrolase/genética , Complexo de Endopeptidases do Proteassoma , Proteínas Proto-Oncogênicas c-akt/fisiologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53
3.
J Pharmacol Exp Ther ; 299(2): 477-82, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11602657

RESUMO

We investigated the possibility that opioids activate a tyrosine kinase (TK) that mediates cardioprotection in an in vivo rat model of myocardial infarction. All animals underwent 30 min of regional ischemia and 2 h of reperfusion. Infarct size was expressed as a percentage of the area at risk (IS/AAR). Control animals had an IS/AAR of 58.2 +/- 0.6. Cardioprotection was induced with the delta1- or delta1/delta2-selective opioid agonists, TAN-67, or D-Ala D-Leu enkephalin (DADLE). Both significantly reduced IS/AAR (28.8 +/- 3.6 and 34.8 +/- 3.8, respectively). The general TK inhibitor, genistein, abolished cardioprotection produced by TAN-67 or DADLE (59.1 +/- 3.2 and 61.5 +/- 3.4, respectively), whereas the structural analog, daidzein, lacking TK inhibitory activity, did not. Interestingly, the selective Src/epidermal growth factor (EGF) receptor TK inhibitor, lavendustin A, did not abolish TAN-67-induced cardioprotection (22.1 +/- 6.8). Similarly, the Src-selective TK antagonist, PP2, had no effect on DADLE-induced cardioprotection (31.1 +/- 7.3). These unexpected findings suggest that Src and EGF receptor TKs are not important in the genesis of cardioprotection produced by TAN-67. Finally, we demonstrate that genistein did not affect protein kinase C (PKC) translocation induced by TAN-67. These data suggest that a TK, but most likely not an Src/EGF receptor TK, is important in cardioprotection via opioid receptor stimulation and that the pathway for TK activation is downstream from or parallel to PKC activation in the in situ rat heart since genistein could not affect PKC translocation of selective isoforms induced by TAN-67 and assessed by immunohistochemistry.


Assuntos
Genes src/fisiologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Proteínas Tirosina Quinases/fisiologia , Receptores Opioides delta/efeitos dos fármacos , Transdução de Sinais/fisiologia , Animais , Imunofluorescência , Hemodinâmica/efeitos dos fármacos , Hemodinâmica/fisiologia , Imuno-Histoquímica , Isoenzimas/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/patologia , Proteína Quinase C/metabolismo , Coelhos , Ratos , Ratos Wistar , Translocação Genética/genética
4.
Am J Physiol Heart Circ Physiol ; 281(3): H1184-92, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11514286

RESUMO

Stress-activated protein kinases may be essential to cardioprotection. We assessed the role of p38 in an in vivo rat model of ischemia-reperfusion. Ischemic preconditioning (IPC) and the delta(1)-opioid receptor agonist 2-methyl-4aalpha-(3-hydroxyphenyl)-1,2,3,4,4a,5,12,12aalpha-octahydroquinolino [2,3,3-g]isoquinoline (TAN-67) significantly reduced infarct size (IS), expressed as a percentage of the area at risk (AAR), versus animals subjected only to 30 min of ischemia and 2 h of reperfusion (7.1 +/- 1.5 and 29.6 +/- 3.3 vs. 59.7 +/- 1.6%). The p38 antagonist SB-203580 attenuated IPC when it was administered before (34.0 +/- 6.9%) or after (25.0 +/- 3.8%) the IPC stimulus; however, it did not significantly attenuate TAN-67-induced cardioprotection (39.6 +/- 3.2). We also assessed the phosphorylation of p38 and c-jun NH(2)-terminal kinase (JNK) throughout ischemia-reperfusion in nuclear and cytosolic fractions. After either intervention, no increase was detected in the phosphorylation state of either enzyme in the nuclear fraction or for p38 in the cytosolic fraction versus control hearts. However, there was a robust increase in JNK activity in the cytosolic fraction immediately on reperfusion that was more pronounced in animals subjected to IPC or administered TAN-67. These data suggest that SB-203580 likely attenuates IPC via the inhibition of kinases other than p38, which may include JNK. The data also suggest that activation of JNK during early reperfusion may be an important component of cardioprotection.


Assuntos
Citoproteção , Precondicionamento Isquêmico Miocárdico , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Isquemia Miocárdica/fisiopatologia , Miocárdio/metabolismo , Animais , Vasos Coronários/fisiopatologia , Vasos Coronários/cirurgia , Citoproteção/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Hemodinâmica/efeitos dos fármacos , Imidazóis/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Isquemia Miocárdica/patologia , Miocárdio/patologia , Fosforilação/efeitos dos fármacos , Piridinas/farmacologia , Quinolinas/farmacologia , Ratos , Ratos Wistar , Receptores Opioides delta/agonistas , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/prevenção & controle , Proteínas Quinases p38 Ativadas por Mitógeno
5.
Basic Res Cardiol ; 96(3): 267-74, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11403420

RESUMO

Ceramide has been shown to be a key signaling molecule involved in the apoptotic effect of tumor necrosis factor alpha (TNF-alpha) and other cytokines. Given the importance of cytokines such as TNF-alpha in myocardial ischemia-reperfusion injury, we hypothesize that ceramide is increased during ischemia or reperfusion, and that the activity of enzymes responsible for its production or breakdown should be increased and/or decreased, respectively. Therefore, in the present study, we characterized the enzymatic activities responsible for ceramide production and metabolism in the myocardium of rats, and determined the contribution of these enzymes to altered ceramide levels during myocardial ischemia and reperfusion. The basal ceramide concentration in the myocardium of rats was 34.0 pmol/mg tissue. As determined by the conversion of 14C-sphingomyelin into ceramide and 14C-choline phosphate, both neutral (N-) and acidic (A-) SMase were detected in the myocardium, with a conversion rate of 0.09 +/- 0.008 and 0.32 +/- 0.05 nmol/min per mg protein, respectively. The activity of A-SMase (78 % of total cellular activity) was significantly higher in microsomes than in cytosol, while the activity of N-SMase was similar in both fractions. Ceramidase, a ceramide-metabolizing enzyme, was also detected in the myocardium of rats. It metabolized ceramide into sphingosine at a rate of 9.94 +/- 0.42 pmol/min per mg protein. In anesthetized rats, 30 min of ischemia had no apparent effect on ceramide concentrations in the myocardium, while 30 min of ischemia followed by 3 h of reperfusion resulted in a significant increase in ceramide by 48 %. The activities of both N- and A-SMase were reduced by 44 % and 32 %, respectively, in the myocardium subjected to ischemia followed by reperfusion, but unaltered in the ischemic myocardium. It was also found that myocardial ischemia followed by reperfusion produced a marked inhibition of ceramidase (by 29 %). These results demonstrate that the myocardium of rats expresses N- and A-SMase and ceramidase, which contribute to the production and metabolism of ceramide, respectively. Tissue ceramide concentrations increased in reperfused myocardium. These increases in ceramide were not associated with enhanced SMase activity, but rather with reduced ceramidase activity.


Assuntos
Ceramidas/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , Animais , Citosol/metabolismo , Modelos Animais de Doenças , Masculino , Microssomos/metabolismo , Ratos , Ratos Wistar , Esfingomielina Fosfodiesterase/metabolismo
6.
Basic Res Cardiol ; 96(2): 136-42, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11327331

RESUMO

Opioids have been previously shown to confer acute and delayed cardioprotection against a prolonged ischemic insult. We have extensively characterized the signal transduction pathway mediating acute cardioprotection and have suggested a role for extracellular signal regulated kinase (ERK) in this cardioprotection. Therefore, we attempted to determine a role for ERK and the stress activated MAP kinase, p38, in opioid-induced delayed cardioprotection by using selective inhibitors of these pathways. All rats were subjected to 30 min of ischemia and 2 h of reperfusion (I/R). Control animals, injected with saline 48 h prior to I/R, had an infarct size/area at risk (IS/AAR) of 61.6 +/- 1.6.48-h pretreatment with TAN-67 (30 mg/kg), a delta1-opioid receptor agonist, maximally reduced IS/AAR (31.2 +/- 6.5). The involvement of ERK was examined with PD 098059, a selective pharmacological antagonist which inhibits the upstream kinase, MEK-1, that phosphorylates and activates ERK. PD 098059 (0.3 mg/kg) did not alter IS/AAR when administered alone (60.7 +/- 4.9). However, PD 098059 (0.3 mg/kg) administration 30 min prior to TAN-67 (30 mg/kg) completely abolished cardioprotection (61.0 +/- 7.6). The selective p38 inhibitor, SB 203580 (1.0 mg/kg), had no effect on IS/AAR in the absence of TAN-67 (53.1 +/- 2.3). Additionally, SB 203580 (1.0 mg/kg) when administered prior to TAN-67 (30 mg/kg) partially abolished cardioprotection (51.3 +/- 6.4). These results suggest that both ERK and p38 are integral components of opioid-induced delayed cardioprotection and may act via parallel pathways.


Assuntos
Precondicionamento Isquêmico Miocárdico , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Isquemia Miocárdica/metabolismo , Receptores Opioides/metabolismo , Analgésicos/farmacologia , Animais , Pressão Sanguínea , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Frequência Cardíaca , Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Isquemia Miocárdica/tratamento farmacológico , Miocárdio/enzimologia , Piridinas/farmacologia , Quinolinas/farmacologia , Ratos , Ratos Wistar , Proteínas Quinases p38 Ativadas por Mitógeno
7.
J Mol Cell Cardiol ; 33(4): 831-4, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11273735

RESUMO

We have previously demonstrated that K(ATP)channel openers administered just prior to and throughout reperfusion induce cardioprotection in the blood-perfused canine heart. However, a recent report suggests that the mitochondrial K(ATP)channel is only a trigger of ischemic preconditioning (IPC). These recent data are, however, in contrast to most previous investigations that suggested that activation of the mitochondrial K(ATP)channel is an important downstream mediator of cardioprotection. Therefore, we examined the role of the mitochondrial K(ATP)channel as a downstream mediator of IPC in a rat model by administering the selective mitochondrial K(ATP)channel antagonist, 5-hydroxydecanoate (5-HD), at several points during IPC. Infarct size (IS) was determined by tetrazolium chloride staining and expressed as a percent of the area at risk (AAR). Control animals had an IS/AAR of 58.4+/-0.6 and IS/AAR was reduced to 6.2+/-1.7 following IPC. 5-HD (10 mg/kg), attenuated cardioprotection when administered either 5 min prior to the IPC stimulus (40.4+/-1.4), during the reperfusion phase of the IPC stimulus (39.7+/-5.9), or 5 min prior to reperfusion during prolonged ischemia (34.3+/-6.9). Additionally, when 5-HD was administered at 5 mg/kg during the reperfusion phase of index ischemia plus 5 min prior to IPC or plus during the reperfusion phase of IPC, cardioprotection was also attenuated (36.3+/-5.5 and 43.8+/-6.9, respectively). These data suggest that activation of the mitochondrial K(ATP) channel is an important downstream regulator of myocardial protection with effects lasting into the reperfusion period following prolonged ischemia.


Assuntos
Trifosfato de Adenosina/metabolismo , Ativação do Canal Iônico/fisiologia , Precondicionamento Isquêmico Miocárdico , Mitocôndrias/metabolismo , Isquemia Miocárdica/fisiopatologia , Reperfusão Miocárdica , Canais de Potássio/fisiologia , Animais , Coração , Hemodinâmica , Masculino , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Canais de Potássio/metabolismo , Ratos , Ratos Wistar
8.
J Pharmacol Exp Ther ; 296(2): 642-9, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11160653

RESUMO

Stimulation of the delta(1)-opioid receptor has been shown to trigger ischemic preconditioning (IPC). Additionally, myocardial ischemia/reperfusion induces the activation of extracellular signal-regulated kinase (ERK). Therefore, we examined the role of ERK in acute cardioprotection induced by delta(1)-opioid receptor stimulation or IPC. Infarct size (IS) was expressed as a percentage of the area at risk (AAR). Control animals had an IS/AAR of 60.6 +/- 1.8. IPC and delta(1)-opioid receptor stimulation with TAN-67 reduced IS/AAR (8.2 +/- 1.3 and 30.2 +/- 2.4). Inhibition of ERK with the selective MEK-1 antagonist, PD 098059 during IPC or TAN-67 administration significantly reduced cardioprotection (41.5 +/- 6.4 and 63.0 +/- 4.8). Western Blot analysis and subsequent densitometry corroborated these observations. Control, TAN-67-, or IPC-treated hearts were harvested after 0, 5, 15, and 30 min of ischemia or 5, 30, and 60 min of reperfusion and separated into cytosolic and nuclear fractions. Both isoforms of ERK (p44 and p42) rapidly increased to greater levels throughout reperfusion in the nuclear fraction of IPC- and opioid-treated versus control rats, however, this increase was not attenuated by PD 098059. Conversely, the rapid activation of the 44-kDa isoform of ERK after 5 min of reperfusion in the cytosolic fraction was significantly increased in IPC- and opioid-treated hearts versus control, and this increase was abolished by pretreatment with PD 098059. Additionally, p42 was activated in the cytosolic fraction of IPC-treated animals. These results suggest a key role for the 44-kDa isoform of ERK in the cytoplasm during cardioprotection induced by either IPC or stimulation of the delta(1)-opioid receptor.


Assuntos
Analgésicos Opioides/uso terapêutico , Cardiopatias/prevenção & controle , Precondicionamento Isquêmico Miocárdico , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Western Blotting , Células COS , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/enzimologia , Citosol/efeitos dos fármacos , Citosol/enzimologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/uso terapêutico , Frequência Cardíaca/efeitos dos fármacos , Isoenzimas/efeitos dos fármacos , Isoenzimas/metabolismo , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Infarto do Miocárdio/patologia , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Ratos , Ratos Wistar , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas
9.
Am J Physiol Heart Circ Physiol ; 280(3): H1346-53, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11179083

RESUMO

Stimulation of the delta(1)-opioid receptor confers cardioprotection to the ischemic myocardium. We examined the role of protein kinase C (PKC) after delta-opioid receptor stimulation with TAN-67 or D-Ala(2)-D-Leu(5)-enkephalin (DADLE) in a rat model of myocardial infarction induced by a 30-min coronary artery occlusion and 2-h reperfusion. Infarct size (IS) was determined by tetrazolium staining and expressed as a percentage of the area at risk (IS/AAR). Control animals, subjected to ischemia and reperfusion, had an IS/AAR of 59.9 +/- 1.8. DADLE and TAN-67 administered before ischemia significantly reduced IS/AAR (36.9 +/- 3.9 and 36.7 +/- 4.7, respectively). The delta(1)-selective opioid antagonist 7-benzylidenenaltrexone (BNTX) abolished TAN-67-induced cardioprotection (54.4 +/- 1.3). Treatment with the PKC antagonist chelerythrine completely abolished DADLE- (61.8 +/- 3.2) and TAN-67-induced cardioprotection (55.4 +/- 4.0). Similarly, the PKC antagonist GF 109203X completely abolished TAN-67-induced cardioprotection (54.6 +/- 6.6). Immunofluorescent staining with antibodies directed against specific PKC isoforms was performed in myocardial biopsies obtained after 15 min of treatment with saline, chelerythrine, BNTX, or TAN-67 and chelerythrine or BNTX in the presence of TAN-67. TAN-67 induced the translocation of PKC-alpha to the sarcolemma, PKC-beta(1) to the nucleus, PKC-delta to the mitochondria, and PKC-epsilon to the intercalated disk and mitochondria. PKC translocation was abolished by chelerythrine and BNTX in TAN-67-treated rats. To more closely examine the role of these isoforms in cardioprotection, we utilized the PKC-delta selective antagonist rottlerin. Rottlerin abolished opioid-induced cardioprotection (48.9 +/- 4.8) and PKC-delta translocation without affecting the translocation of PKC-alpha, -beta(1), or -epsilon. These results suggest that PKC-delta is a key second messenger in the cardioprotective effects of delta(1)-opioid receptor stimulation in rats.


Assuntos
Leucina Encefalina-2-Alanina/farmacologia , Precondicionamento Isquêmico Miocárdico , Isoenzimas/metabolismo , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/metabolismo , Naltrexona/análogos & derivados , Proteína Quinase C/metabolismo , Acetofenonas/farmacologia , Alcaloides , Analgésicos/farmacologia , Animais , Benzofenantridinas , Benzopiranos/farmacologia , Compostos de Benzilideno/farmacologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Frequência Cardíaca , Indóis/farmacologia , Masculino , Maleimidas/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/enzimologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Fenantridinas/farmacologia , Proteína Quinase C-delta , Quinolinas/farmacologia , Ratos , Ratos Wistar , Receptores Opioides delta/agonistas , Receptores Opioides delta/antagonistas & inibidores
10.
J Pharmacol Exp Ther ; 294(2): 451-7, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10900218

RESUMO

We examined the role of the sarcolemmal and mitochondrial ATP-sensitive potassium (K(ATP)) channel in a rat model of myocardial infarction after stimulation with the selective delta(1)-opioid receptor agonist TAN-67. Hearts were subjected to 30 min of regional ischemia and 2 h of reperfusion. Infarct size was expressed as a percentage of the area at risk. TAN-67 significantly reduced infarct size/area at risk (29.6 +/- 3.3) versus control (63. 1 +/- 2.3). The sarcolemmal-selective K(ATP) channel antagonist HMR 1098, administered 10 min before TAN-67, did not significantly attenuate cardioprotection (26.0 +/- 7.3) at a dose (3 mg/kg) that had no effect in the absence of TAN-67 (56.3 +/- 4.3). Pretreatment with the mitochondrial selective antagonist 5-hydroxydecanoic acid (5-HD) 5 min before the 30-min occlusion completely abolished TAN-67-induced cardioprotection (54.3 +/- 2.7), but had no effect in the absence of TAN-67 (62.6 +/- 4.1), suggesting the involvement of the mitochondrial K(ATP) channel. Additionally, we examined the antiarrhythmic effects of TAN-67 in the presence or absence of 5-HD and HMR 1098 during 30 min of ischemia. Control animals had an average arrhythmia score of 10.40 +/- 2.41. TAN-67 significantly reduced the arrhythmia score during 30 min of ischemia (2.38 +/- 0. 85). 5-HD and HMR 1098 in the absence of TAN-67 produced an insignificant decrease in the arrhythmia score (8.80 +/- 2.56 and 4. 20 +/- 1.07, respectively). 5-HD administration before TAN-67 treatment abolished its antiarrhythmic effect (4.71 +/- 1.11). However, HMR 1098 did not abolish TAN-67-induced protection against arrhythmias (1.67 +/- 0.80). These data suggest that delta(1)-opioid receptor stimulation is cardioprotective against myocardial ischemia and sublethal arrhythmias and suggest a role for the mitochondrial K(ATP) channel in mediating these cardioprotective effects.


Assuntos
Analgésicos Opioides/uso terapêutico , Antiarrítmicos/farmacologia , Arritmias Cardíacas/prevenção & controle , Mitocôndrias Cardíacas/efeitos dos fármacos , Infarto do Miocárdio/prevenção & controle , Bloqueadores dos Canais de Potássio , Quinolinas/uso terapêutico , Sarcolema/efeitos dos fármacos , Trifosfato de Adenosina/fisiologia , Analgésicos Opioides/antagonistas & inibidores , Animais , Antiarrítmicos/antagonistas & inibidores , Benzamidas/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Ácidos Decanoicos/farmacologia , Interações Medicamentosas , Frequência Cardíaca/efeitos dos fármacos , Hidroxiácidos/farmacologia , Masculino , Mitocôndrias Cardíacas/fisiologia , Isquemia Miocárdica/complicações , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Quinolinas/antagonistas & inibidores , Ratos , Ratos Wistar , Receptores Opioides delta/agonistas , Sarcolema/fisiologia , Fibrilação Ventricular/prevenção & controle
11.
Am J Physiol Heart Circ Physiol ; 278(1): H305-12, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10644614

RESUMO

We examined the role of the sarcolemmal and mitochondrial K(ATP) channels in a rat model of ischemic preconditioning (IPC). Infarct size was expressed as a percentage of the area at risk (IS/AAR). IPC significantly reduced infarct size (7 +/- 1%) versus control (56 +/- 1%). The sarcolemmal K(ATP) channel-selective antagonist HMR-1098 administered before IPC did not significantly attenuate cardioprotection. However, pretreatment with the mitochondrial K(ATP) channel-selective antagonist 5-hydroxydecanoic acid (5-HD) 5 min before IPC partially abolished cardioprotection (40 +/- 1%). Diazoxide (10 mg/kg iv) also reduced IS/AAR (36.2 +/- 4.8%), but this effect was abolished by 5-HD. As an index of mitochondrial bioenergetic function, the rate of ATP synthesis in the AAR was examined. Untreated animals synthesized ATP at 2.12 +/- 0.30 micromol x min(-1) x mg mitochondrial protein(-1). Rats subjected to ischemia-reperfusion synthesized ATP at 0.67 +/- 0.06 micromol x min(-1) x mg mitochondrial protein(-1). IPC significantly increased ATP synthesis to 1.86 +/- 0.23 micromol x min(-1) x mg mitochondrial protein(-1). However, when 5-HD was administered before IPC, the preservation of ATP synthesis was attenuated (1.18 +/- 0.15 micromol x min(-1) x mg mitochondrial protein(-1)). These data are consistent with the notion that inhibition of mitochondrial K(ATP) channels attenuates IPC by reducing IPC-induced protection of mitochondrial function.


Assuntos
Precondicionamento Isquêmico Miocárdico , Proteínas de Membrana/fisiologia , Mitocôndrias Cardíacas/fisiologia , Trifosfato de Adenosina/biossíntese , Trifosfato de Adenosina/metabolismo , Animais , Ácidos Decanoicos/farmacologia , Diazóxido/farmacologia , Hemodinâmica/efeitos dos fármacos , Hidroxiácidos/farmacologia , Masculino , Proteínas de Membrana/antagonistas & inibidores , Mitocôndrias Cardíacas/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Concentração Osmolar , Canais de Potássio , Ratos , Ratos Wistar
12.
Circ Res ; 84(7): 846-51, 1999 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-10205153

RESUMO

Opioids have been previously shown to confer short-term cardioprotection against a prolonged ischemic insult. Therefore, the present study was designed to determine whether opioids can induce a delayed or "second window" of cardioprotection and to assess the potential involvement of the mitochondrial KATP channel. All rats were subjected to 30 minutes of ischemia and 2 hours of reperfusion (I/R). Control animals, injected with saline 24 hours before I/R, elicited an infarct size/area at risk (IS/AAR) of 62.9+/-3.4. TAN-67, a delta1-opioid receptor agonist, was administered 10 or 30 mg/kg IP 12, 24, 48, or 72 hours before I/R. TAN-67 (10 mg/kg) 12- or 24-hour pretreatment did not significantly reduce IS/AAR (62.1+/-6.3 and 43.3+/-7.3, respectively). Similarly, 12-hour pretreatment with TAN-67 (30 mg/kg) did not reduce IS/AAR (60.0+/-5.6); however, 24-hour pretreatment significantly reduced IS/AAR (34.5+/-5.9). Forty-eight-hour pretreatment with TAN-67 maximally reduced IS/AAR (29.2+/-7.0), and opioid-induced cardioprotection was lost after 72-hour pretreatment (61.7+/-3.8). TAN-67-induced cardioprotection could be abolished by pretreatment with the selective delta1-opioid receptor antagonist 7-benzylidenenaltrexone, BNTX, administered either 30 minutes before TAN-67 given 48 hours before I/R or 10 minutes before I/R in rats previously treated for 48 hours with TAN-67 (59.6+/-3.1 and 58.7+/-3.5, respectively). The involvement of the KATP channel was investigated with 2 inhibitors: glibenclamide, a nonselective KATP channel inhibitor, and 5-hydroxydecanoic acid, selective for the mitochondrial KATP channel in rabbits. Glibenclamide, administered 30 minutes before I/R in 48-hour TAN-67-pretreated rats, completely abolished cardioprotection (60. 4+/-3.2). Similarly, 5-hydroxydecanoic acid, administered 5 minutes before I/R in rats pretreated 48 hours previously with TAN-67, completely abolished cardioprotection (57.8+/-2.5). These results suggest that delta1-opioid receptor stimulation, 24 to 48 hours before an ischemic insult, produces a delayed cardioprotective effect that is possibly the result of mitochondrial KATP channel activation.


Assuntos
Mitocôndrias/química , Isquemia Miocárdica/fisiopatologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Canais de Potássio/fisiologia , Receptores Opioides delta/fisiologia , Analgésicos/farmacologia , Animais , Compostos de Benzilideno/farmacologia , Pressão Sanguínea , Circulação Coronária , Glibureto/farmacologia , Hipoglicemiantes/farmacologia , Precondicionamento Isquêmico Miocárdico , Masculino , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/fisiopatologia , Isquemia Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Miocárdio/química , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Quinolinas/farmacologia , Ratos , Ratos Wistar
13.
Am J Physiol ; 276(4): H1229-35, 1999 04.
Artigo em Inglês | MEDLINE | ID: mdl-10199847

RESUMO

Both tyrosine kinase (TK) and protein kinase C (PKC) inhibitors have been shown individually to completely abolish the cardioprotective effects of ischemic preconditioning (IPC) in rabbits; however, blockade of both enzymes is necessary to totally abolish IPC in pigs. Recently, we have shown that TK inhibition partially attenuates the cardioprotective effect of IPC in intact rat hearts. Therefore, the present study was designed to test the hypothesis that inhibition of both TK and PKC is necessary to completely abolish IPC in the intact rat and that this effect is dependent on the intensity of the preconditioning stimulus. All animals were subjected to 30 min of coronary artery occlusion and 2 h of reperfusion. In series 1, multiple-cycle-induced IPC was produced via three 5-min occlusions interspersed with 5 min of reperfusion (3 x 5 IPC). Genistein (5 mg/kg), a TK inhibitor infused 30 min before IPC, and chelerythrine chloride (5 mg/kg), a PKC inhibitor infused 5 min before the prolonged ischemic insult, were administered alone or in combination in the absence or presence of 3 x 5 IPC. 3 x 5 IPC produced a marked reduction in infarct size as a percentage of area at risk compared with control (8.0 +/- 0.8 vs. 56.1 +/- 0.8%). The effects of 3 x 5 IPC were partially blocked by pretreatment with genistein (34.0 +/- 2.0%) or chelerythrine (26.4 +/- 2.8%) alone; however, combined administration of genistein and chelerythrine completely abolished the effects of 3 x 5 IPC (50.7 +/- 3.6%). In series 2, single-cycle IPC was elicited by one 5-min occlusion followed by 10 min of reperfusion (1 x 5 IPC). Compared with control, 1 x 5 IPC also significantly reduced infarct size (15.4 +/- 3.0%). Genistein or chelerythrine administered alone completely abolished 1 x 5 IPC-induced cardioprotection. These results suggest that the efficacy of TK and PKC inhibition to block IPC depends on the intensity of the preconditioning stimulus and that these kinases may work through parallel pathways.


Assuntos
Precondicionamento Isquêmico Miocárdico , Proteína Quinase C/fisiologia , Proteínas Tirosina Quinases/fisiologia , Alcaloides , Animais , Benzofenantridinas , Combinação de Medicamentos , Inibidores Enzimáticos/farmacologia , Genisteína/farmacologia , Coração/efeitos dos fármacos , Hemodinâmica/fisiologia , Masculino , Infarto do Miocárdio/patologia , Fenantridinas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Ratos , Ratos Wistar
14.
Am J Physiol ; 275(6): H2009-15, 1998 12.
Artigo em Inglês | MEDLINE | ID: mdl-9843799

RESUMO

Ischemic preconditioning (IPC) confers cardioprotection against a prolonged ischemic insult. Tyrosine kinase (TK) inhibitors have been shown to attenuate IPC; however, it is unclear whether TK is involved in the initiation of and/or the maintenance of this phenomenon. Thus the hypothesis that TK acts primarily during the initiation of IPC was examined in a rat model of myocardial infarction. Hearts were subjected to 30 min of coronary artery occlusion and 2 h of reperfusion. IPC was elicited by three 5-min occlusions interspersed with 5 min of reperfusion before the prolonged occlusion period. Genistein, a nonspecific TK inhibitor, was administered before or during the final 2 min of the first or third occlusion period of IPC. Daidzein, an inactive structural analog of genistein, and lavendustin A, a more specific TK inhibitor, were also tested in this model. IPC markedly reduced infarct size expressed as a percentage of the area at risk compared with control (56.3 +/- 2.8 to 7.1 +/- 2.0%). This cardioprotection was attenuated by genistein pretreatment (5 mg/kg: 34.7 +/- 2.2%, 10 mg/kg: 33.5 +/- 5.9%). However, genistein administered during the first or third occlusion period of IPC did not significantly attenuate cardioprotection (10.3 +/- 2.9% and 6.4 +/- 2.0%). Lavendustin A (1. 0 mg/kg) pretreatment also attenuated IPC (30.1 +/- 2.2%), whereas daidzein (5 mg/kg) had no effect (7.9 +/- 2.4%). These results suggest that activation of a TK is involved in the initiation but not the maintenance of IPC in the rat myocardium.


Assuntos
Inibidores Enzimáticos/farmacologia , Coração/efeitos dos fármacos , Precondicionamento Isquêmico Miocárdico , Proteínas Tirosina Quinases/antagonistas & inibidores , Animais , Genisteína/farmacologia , Hemodinâmica/efeitos dos fármacos , Isoflavonas/farmacologia , Masculino , Infarto do Miocárdio/patologia , Miocárdio/patologia , Fenóis/farmacologia , Ratos , Ratos Wistar , Fatores de Risco
15.
J Mol Cell Cardiol ; 30(9): 1817-25, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9769237

RESUMO

Recent results have shown that the sulfonylurea receptor couples to several types of inward-rectifier potassium (KIR) channels, which suggests that sensitivity to blockade of a pathophysiological phenomenon such as ischemic preconditioning (PC) by glibenclamide may not be the result of this compound selectively blocking the ATP-sensitive potassium (KATP) channel. Therefore, to address this possibility, a role for myocardial KIR v KATP channels in ischemic PC was evaluated in the rat. To test this hypothesis, anesthetized, open-chest, male Wistar rats were assigned to one of seven experimental protocols. Animals assigned to group I (control) received 30 min of occlusion and 2 h of reperfusion. Ischemic PC was produced by 3x5-min occlusion and 2-h reperfusion periods (group II). Terikalant (TK), an inward-rectifier potassium channel blocker, was used to test the role of other K+ channels, most notably the KIR, in the cardioprotective effect of ischemic PC in the rat. TK was given at a dose of 3 mg/kg, i.v., 15 min before the prolonged occlusion and reperfusion periods (group III). In groups IV, V, and VI terikalant (1, 3 and 6 mg/kg, i.v.) was given 15 min before ischemic PC (lowTK+PC, medTK+PC and hiTK+PC, respectively). Group VII consisted of glibenclamide (0.3 mg/kg, i.v.) given 30 min prior to ischemic PC (GLY+PC). Infarct size (IS) as a percent of the area at risk (AAR) was measured using the histochemical stain, 2,3, 5-triphenyltetrazolium chloride. The average IS/AAR for the control was 49.9+/-2.1%. Ischemic PC markedly reduced infarct size (8.6+/-1. 8%; * P<0.05 v control). Terikalant (TK; 1, 3 and 6 mg/kg, i.v.) did not abolish the cardioprotective effect of ischemic PC at any dose (15.5+/-6.4, 16.4+/-5.2 and 8.8+/-1.6%, respectively; * P<0.05 v control). TK itself had no effect on infarct size. GLY completely abolished the cardioprotective effect of ischemic PC (48.2+/-6.4%). In addition, the high dose of TK significantly (P<0.05) increased the action potential duration at 50% repolarization from 48+/-3 to 64+/-4 ms and 30 microM of TK, a concentration which produced a 39% decrease in the inward-rectifier potassium channel current in isolated guinea-pig ventricular myocytes in the whole-cell patch-clamp mode did not block the increase in K ATP current produced by the KATP opener bimakalim (3 microM). These results demonstrate that although the myocardial KATP channel belongs to the K IR superfamily, the endogenous myocardial KIR channel does not mediate ischemic PC in the rat heart; however, the K ATP channel does mediate its cardioprotective effect.


Assuntos
Antiarrítmicos/farmacologia , Cromanos/farmacologia , Coração/fisiologia , Precondicionamento Isquêmico Miocárdico , Piperidinas/farmacologia , Canais de Potássio/fisiologia , Transportadores de Cassetes de Ligação de ATP , Potenciais de Ação , Animais , Células Cultivadas , Eletrofisiologia , Cobaias , Coração/efeitos dos fármacos , Hemodinâmica , Canais KATP , Masculino , Infarto do Miocárdio/fisiopatologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização , Ratos , Ratos Wistar
16.
Am J Physiol ; 275(2): H495-500, 1998 08.
Artigo em Inglês | MEDLINE | ID: mdl-9683437

RESUMO

It has been previously demonstrated that Gi/o proteins are involved in ischemic preconditioning (IPC) in rabbits and dogs; however, there has been controversy as to the role of Gi/o proteins in IPC in in vivo rat infarct models. Therefore, the role of G proteins in the cardioprotective effect of IPC in a rat infarct model was reevaluated. Cardioprotection as indicated by infarct size (IS) as a percentage of the area at risk (IS/AAR) was determined by triphenyltetrazolium stain. The control group, which was subjected to 30 min of occlusion (Occ) and 2 h of reperfusion (Rep), had an IS/AAR of 46 +/- 6%. A single 5-min Occ followed by 10 min of Rep (1x PC) as well as three 5-min Occ periods interspersed with 5 min of Rep (3x PC) markedly reduced IS/AAR (6 +/- 1 and 8 +/- 1%, respectively). Pretreatment with pertussis toxin (10 microg/kg ip) for 48 h before 1x PC or 3x PC completely abolished their cardioprotective effects (46 +/- 5 and 38 +/- 4%, respectively). Pertussis toxin had no effect on IS/AAR and did not inactivate Gi/o proteins as assessed by an in vitro ADP-ribosylation assay of heart homogenates. These results demonstrate that the cardioprotective effect of IPC is mediated by a small subpopulation of Gi/o proteins in the intact rat heart.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Precondicionamento Isquêmico Miocárdico , Infarto do Miocárdio/prevenção & controle , Isquemia Miocárdica/fisiopatologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Toxina Pertussis , Fatores de Virulência de Bordetella/farmacologia , Acetilcolina/farmacologia , Adenosina/farmacologia , Adenosina Difosfato Ribose/metabolismo , Animais , Cães , Frequência Cardíaca/efeitos dos fármacos , Masculino , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , NAD/metabolismo , Coelhos , Ratos , Ratos Wistar
17.
Circulation ; 97(13): 1282-9, 1998 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-9570199

RESUMO

BACKGROUND: Our laboratory has previously shown that delta-opioid receptors are involved in the cardioprotective effect of ischemic preconditioning in the rat heart. However, this class of receptors consists of two subtypes, delta1, and delta2, and mu- or kappa-opioid receptors may also exist in the heart. Therefore, the purpose of the present study was to test the hypothesis that ischemic preconditioning is mediated through stimulation of one or both delta-opioid receptor subtypes. METHODS AND RESULTS: Anesthetized, open chest, male Wistar rats were assigned to 1 of 14 groups. All animals were subjected to 30 minutes of occlusion and 2 hours of reperfusion. Ischemic preconditioning was elicited by three 5-minute occlusion periods interspersed with 5 minutes of reperfusion. Two doses of 7-benzylidenenaltrexone (BNTX; 1 and 3 mg/kg i.v.), a selective delta1-opioid receptor antagonist, or naltriben (NTB; 1 and 3 mg/kg i.v.), a selective delta2-opioid receptor antagonist, were given before ischemic preconditioning. To test for a role of mu-opioid receptors, rats were pretreated with beta-funaltrexamine (beta-FNA; 15 mg/kg s.c), an irreversible mu-opioid receptor antagonist, 24 hours before ischemic preconditioning or given the mu-opioid receptor agonist D-Ala,2N-Me-Phe,4glycerol5-enkephalin (DAMGO) as three 5-minute infusions (1, 10, and 100 microg/kg per infusion i.v., respectively) interspersed with 5-minute drug-free periods before the prolonged ischemic and reperfusion periods (lowDAMGO, medDAMGO, and hiDAMGO, respectively). The involvement of kappa-opioid receptors was tested by administering one of two doses of nor-binaltorphimine (nor-BNI; 1 and 5 mg/kg i.v.) before ischemic preconditioning. Infarct size (IS) as a percent of the area at risk (AAR) was measured by triphenyltetrazolium stain. Ischemic preconditioning markedly reduced IS/AAR (14+/-4%, P<.05) compared with control (55+/-4%). NTB, beta-FNA, and nor-BNI were unable to block the cardioprotective effect of ischemic preconditioning. In addition, DAMGO had no effect on IS/AAR. However, the high dose of BNTX (3 mg/kg i.v.) significantly attenuated the cardioprotective effect of ischemic preconditioning (39+/-5%; P<.05 versus control and ischemic preconditioning). CONCLUSIONS: These results indicate that delta1-opioid receptors play an important role in the cardioprotective effect of ischemic preconditioning in the rat heart.


Assuntos
Precondicionamento Isquêmico Miocárdico , Receptores Opioides delta/fisiologia , Receptores Opioides kappa/fisiologia , Receptores Opioides mu/fisiologia , Animais , Compostos de Benzilideno/uso terapêutico , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/uso terapêutico , Hemodinâmica/efeitos dos fármacos , Masculino , Infarto do Miocárdio/tratamento farmacológico , Naltrexona/análogos & derivados , Naltrexona/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Ratos , Ratos Wistar , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos
18.
Am J Physiol ; 274(3): H909-14, 1998 03.
Artigo em Inglês | MEDLINE | ID: mdl-9530203

RESUMO

We have previously shown that delta (delta)-opioid receptors, most notably delta 1, are involved in the cardioprotective effect of ischemic preconditioning (PC) in rats; however, the mechanism by which delta-opioid receptor-induced cardioprotection is mediated remains unknown. Therefore, we hypothesized that several of the known mediators of ischemic PC such as the ATP-sensitive potassium (KATP) channel and Gi/o proteins are involved in the cardioprotective effect produced by delta 1-opioid receptor activation. To address these possibilities, anesthetized, open-chest Wistar rats were randomly assigned to five groups. Control animals were subjected to 30 min of coronary artery occlusion and 2 h of reperfusion. To demonstrate that stimulating delta 1-opioid receptors produces cardioprotection, TAN-67, a new selective delta 1-agonist, was infused for 15 min before the long occlusion and reperfusion periods. In addition, one group received 7-benzylidenenaltrexone (BNTX), a selective delta 1-antagonist, before TAN-67. To study the involvement of KATP channels or Gi/o proteins in delta 1-opioid receptor-induced cardioprotection, glibenclamide (Glib), a KATP channel antagonist, or pertussis toxin (PTX), an inhibitor of Gi/o proteins, was administered before TAN-67. Infarct size (IS) as a percentage of the area at risk (IS/AAR) was determined by tetrazolium stain. TAN-67 significantly reduced IS/AAR as compared with control (56 +/- 2 to 27 +/- 5%, n = 5, P < 0.05). The cardioprotective effect of TAN-67 was completely abolished by BNTX, Glib, and PTX (51 +/- 3, 53 +/- 5, and 61 +/- 4%, n = 6 for each group, respectively). These results are the first to suggest that stimulating the delta 1-opioid receptor elicits a cardioprotective effect that is mediated via Gi/o proteins and KATP channels in the intact rat heart.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Infarto do Miocárdio/patologia , Canais de Potássio/fisiologia , Quinolinas/farmacologia , Receptores Opioides delta/agonistas , Animais , Glicemia/metabolismo , Pressão Sanguínea , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/antagonistas & inibidores , Frequência Cardíaca , Precondicionamento Isquêmico Miocárdico , Masculino , Infarto do Miocárdio/tratamento farmacológico , Toxina Pertussis , Ratos , Ratos Wistar , Fatores de Virulência de Bordetella/farmacologia
19.
J Mol Cell Cardiol ; 29(8): 2187-95, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9281450

RESUMO

Several investigators have demonstrated that the opioid pathway is involved in tissue preservation during hypoxia or ischemia and that this protection is mediated via the delta (delta)-opioid receptor. Subsequently, we have shown that opioid receptors are involved in ischemic preconditioning (PC) in the rat heart and that morphine produces a cardioprotective effect; however, the class of opioid receptors involved in mediating these effects is still unknown. Therefore, the purpose of the present study was to test the hypothesis that ischemia- and morphine-induced cardioprotection are mediated via stimulation of the delta-opioid receptor in the rat heart. Anesthetized, open-chest Wistar rats were subjected to one of six protocols. The control group was subjected to 30 min of occlusion and 2 h of reperfusion. Ischemic PC was elicited by three 5 min occlusion periods interspersed with 5 min of reperfusion. Morphine-induced cardioprotection was produced by three 5 min morphine infusions (100 microg/kg/infusion, i.v.) interspersed with a 5-min drug-free period. To determine if the delta-opioid receptor has a role in ischemic PC and morphine-induced cardioprotection, naltrindole (NTI), a selective delta-opioid receptor antagonist, was utilized. NTI (5 mg/kg, i.v.) was given 10 min prior to ischemic PC (NTI+PC) or morphine infusion (NTI+MOR). Also, NTI (5 mg/kg, i.v.) was given 10 min before the 30 min occlusion period in untreated rats. Infarct size (IS) as a percent of the area at risk (AAR) was determined by 2,3,5-triphenyltetrazolium chloride staining. Ischemic PC and morphine infusions resulted in similar reductions in IS/AAR from 51+/-4 to 11+/-3 and 15+/-4% (*P<0.05), respectively. NTI completely abolished the cardioprotective effect induced by ischemia and morphine. The results of the present study suggests a role of delta;-opioid receptors in ischemic PC or morphine-induced myocardial protection in the rat.


Assuntos
Fármacos Cardiovasculares/farmacologia , Coração/efeitos dos fármacos , Precondicionamento Isquêmico Miocárdico , Morfina/farmacologia , Miocárdio/metabolismo , Receptores Opioides delta/fisiologia , Animais , Fármacos Cardiovasculares/uso terapêutico , Masculino , Morfina/uso terapêutico , Infarto do Miocárdio/patologia , Infarto do Miocárdio/prevenção & controle , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Wistar , Receptores Opioides delta/efeitos dos fármacos , Tiopental/análogos & derivados , Tiopental/farmacologia
20.
J Mol Cell Cardiol ; 29(5): 1355-62, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9201621

RESUMO

Previously, our laboratory has shown that opioid receptors are involved in ischemic preconditioning (PC) in the intact rat heart; however, it is not known whether this cardioprotection is mediated by central or peripheral mechanisms. To test this hypothesis, both naloxone (NL), the non-selective opioid receptor antagonist and naloxone methiodide (QNL), its quaternary derivative which does not cross the blood-brain barrier, were used to determine if opioid receptor-induced myocardial protection occurs via a central or peripheral locus of action in inactin-anesthetized, open-chested, Wistar rats. In group I, the control group was subjected to 30 min of occlusion and 2 h of reperfusion. In group II, ischemic PC was elicited by three 5-min occlusion periods interspersed with 5 min of reperfusion. In group III, QNL (10 mg/kg, i.v.) was administered 10 min before the 30 min of occlusion. Groups IV and V consisted of a dose-response effect of QNL on ischemic PC in which QNL (0.3 or 10 mg/kg, i.v., respectively) was given 10 min prior to ischemic PC. In addition, in groups VI and VII, one of two doses of naloxone (1 or 3 mg/kg, i.v.) was administered 10 min before ischemic PC. Infarct size (IS) as a percentage of the area at risk (AAR) was determined by tetrazolium staining. Ischemic PC reduced IS to 9 +/- 2% (P < 0.05) v control (53 +/- 4%). The low dose of QNL partially blocked the cardioprotective effect of ischemic PC; whereas the high dose completely abolished its cardioprotective effect. The high dose of QNL had no effect on IS alone. Similarly, the low dose of NL did not antagonize the cardioprotective effect of ischemic PC; however, the high dose completely abolished ischemic PC. These results indicate that the cardioprotective effect of ischemic preconditioning is mediated by a peripheral opioid receptor mechanism in the intact rat heart.


Assuntos
Precondicionamento Isquêmico Miocárdico , Miocárdio/metabolismo , Receptores Opioides/metabolismo , Animais , Coração , Hemodinâmica , Masculino , Estrutura Molecular , Infarto do Miocárdio , Naloxona/análogos & derivados , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Compostos de Amônio Quaternário , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...