Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Lasers Surg Med ; 42(6): 473-80, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20662023

RESUMO

BACKGROUND AND OBJECTIVE: Light in the visible and near infrared region stimulates various cellular processes, and thus has been used for therapeutic purposes. One of the proposed mechanisms is based on cellular production of reactive oxygen species (ROS) in response to illumination. In the present study, we followed visible light (VL)-induced hydroxyl radicals in various cell types and cellular sites using the electron paramagnetic resonance (EPR) spin-trapping technique. MATERIALS AND METHODS: Fibroblasts, sperm cells, cardiomyocytes, and skeletal muscle cells were irradiated with broadband (400-800 nm) VL. To detect ROS, the EPR spin-trapping technique coupled with the spin-traps 5,5-dimethyl pyrroline-N-oxide (DMPO) or 5-(diethoxyphosphoryl)-5-methyl-1-pyrroline-N-oxide (DEPMPO) were used. To investigate the cellular sites of ROS formation, the cell-permeable molecule, isopropanol, or the nonpermeable proteins, bovine serum albumin (BSA) and superoxide dismutase (SOD), were introduced to the cells before irradiation. ROS production in mitochondria was measured using the fluorescent probe, MitoTracker Red (MTR). RESULTS AND CONCLUSIONS: The concentration of .OH increased both with illumination time and with cell concentration, and decreased when N(2) was bubbled into the cell culture, suggesting that VL initiates a photochemical reaction via endogenous photosensitizers. VL was found to stimulate ROS generation both in membrane and cytoplasm. In addition, fluorescent measurments confirmed the mitochondria to be target for light-cell interaction. The findings support the hypothesis that ROS are generated in various cellular sites following light illumination.


Assuntos
Fibroblastos/metabolismo , Luz , Músculo Esquelético/citologia , Miócitos Cardíacos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Espermatozoides/metabolismo , Animais , Bovinos , Linhagem Celular , Membrana Celular/metabolismo , Óxidos N-Cíclicos , Citoplasma/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Fluorescência , Radical Hidroxila/metabolismo , Masculino , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Pirróis , Ratos
2.
Mol Cell Biochem ; 283(1-2): 75-83, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16444588

RESUMO

Delta-9-tetrahydrocannabinol (THC), the major active component of marijuana, has a beneficial effect on the cardiovascular system during stress conditions, but the defence mechanism is still unclear. The present study was designed to investigate the central (CB1) and the peripheral (CB2) cannabinoid receptor expression in neonatal cardiomyoctes and possible function in the cardioprotection of THC from hypoxia. Pre-treatment of cardiomyocytes that were grown in vitro with 0.1 - 10 microM THC for 24 h prevented hypoxia-induced lactate dehydrogenase (LDH) leakage and preserved the morphological distribution of alpha-sarcomeric actin. The antagonist for the CB2 (10 microM), but not CB1 receptor antagonist (10 microM) abolished the protective effect of THC. In agreement with these results using RT-PCR, it was shown that neonatal cardiac cells express CB2, but not CB1 receptors. Involvement of NO in the signal transduction pathway activated by THC through CB2 was examined. It was found that THC induces nitric oxide (NO) production by induction of NO synthase (iNOS) via CB2 receptors. L-NAME (NOS inhibitor, 100 microM) prevented the cardioprotection provided by THC. Taken together, our findings suggest that THC protects cardiac cells against hypoxia via CB2 receptor activation by induction of NO production. An NO mechanism occurs also in the classical pre-conditioning process; therefore, THC probably pre-trains the cardiomyocytes to hypoxic conditions.


Assuntos
Analgésicos não Narcóticos/uso terapêutico , Dronabinol/uso terapêutico , Hipóxia/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Óxido Nítrico/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Actinas/metabolismo , Animais , Animais Recém-Nascidos , Glioma/tratamento farmacológico , Glioma/metabolismo , L-Lactato Desidrogenase/metabolismo , Miócitos Cardíacos/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Receptor CB1 de Canabinoide/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
3.
Life Sci ; 78(10): 1098-102, 2006 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-16202428

RESUMO

Leptin, a circulating hormone mainly produced by adipose tissue, regulates fatty acid metabolism and causes multiple systemic biological actions even the regulation of cardiovascular function. It is previously known that leptin is a hypoxia-inducible hormone, that hypoxic conditions increase the expression of this peptide in various tissues such as placenta, pancreas and also in the heart. Since leptin receptors are present in the heart, we hypothesized that whether leptin was a protector response for tissues especially for the heart against the deleterious effects of hypoxia. Cultured cardiomyocytes from newborn rats were initially treated with 3000 ng/ml leptin incubation for 1, 5 and 20 h separately, then subjected to 120 min of hypoxia. Hypoxic damage of myocytes was assayed using the measurements of both lactate dehydrogenase and creatine kinase releases into the medium and performing morphological observations (ultrastructural and immunocytochemical) of plates. The obtained results from leptin treated and non-treated control groups were compared to each other, and these data have demonstrated that 5 h of leptin treatment before hypoxia provides a significant protection for cardiomyocytes against hypoxia. Neither 1- nor 20-h leptin treated groups exhibited sufficient protection against hypoxia. In conclusion, leptin protects the cardiomyocyte cultures from hypoxia, but this effect is selective and evident only in the 5-h treated myocytes.


Assuntos
Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Leptina/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Animais , Células Cultivadas , Creatina Quinase/sangue , Meios de Cultura , Desmina/farmacologia , Imuno-Histoquímica , L-Lactato Desidrogenase/sangue , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/ultraestrutura , Ratos
4.
J Mol Cell Cardiol ; 39(1): 149-58, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15893762

RESUMO

Mitochondrial disorder is characteristic of many myocardial injuries such as endotoxemia, shock, acidosis, ischemia/reperfusion, and others. The goal of possible therapy is to increase ATP production. Derivatives of vitamins K may be a potent electron carrier between various mitochondrial electron-donating and electron-accepting enzyme complexes. We aimed to test the possibility that menadione or its water-soluble derivative AK-135, the newly synthesized analogues of vitamin K1--N-derivatives of 2-methyl-3-aminomethyl 1.4-naphthoquinone, would reduce cardiomyocyte damage after hypoxia or mitochondrial respiratory chain inhibition in culture. Menadione, and more effectively, AK-135, restored the electron flow in defective respiratory chain (hypoxia or rotenone) systems. As was shown in this study, 3 microM of AK-135 restored ATP production after blockade of electron flow through mitochondrial complex I with 5 microM rotenone up to 13.18+/-1.56 vs. 3.21+/-1.12 nmol/mg protein in cells treated with rotenone only. In cultures pretreated with 4 microM dicumarol (DT-diaphorase inhibitor), the protective effect of AK-135 and menadione was abolished completely (1.67+/-1.43 and 2.97+/-0.57 nmol/mg protein, respectively). Inhibition of mitochondrial oxidative phosphorylation caused an increase in intracellular Ca(2+) levels. Here we have demonstrated restoration of calcium oscillations and cardiomyocyte contractility by menadione and its derivative after blockade of NADH: ubiquinone oxidoreductase with rotenone, and decrease of Ca(2+) overloading during hypoxia.


Assuntos
Doenças Mitocondriais/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Vitamina K 3/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/metabolismo , Hipóxia Celular , Células Cultivadas , L-Lactato Desidrogenase/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Doenças Mitocondriais/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , NAD(P)H Desidrogenase (Quinona)/antagonistas & inibidores , NAD(P)H Desidrogenase (Quinona)/efeitos dos fármacos , NAD(P)H Desidrogenase (Quinona)/metabolismo , Naftoquinonas/farmacologia , Ratos , Rotenona/farmacologia , Desacopladores/farmacologia , Vitamina K 3/análogos & derivados
5.
Biochem Pharmacol ; 69(8): 1215-23, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15794942

RESUMO

Cardiomyocytes express one or more subtypes of P2 receptors for extracellular nucleotides. P2 purinoceptors, which are activated by nucleotides, are classified as P2X or P2Y: P2X receptors are ligand-gated intrinsic ion channels, and P2Y receptors are G protein-coupled receptors. Extracellular pyrimidine and purine nucleotides are released from the heart during hypoxia. Although the cardioprotective effects of purines acting via purinoceptors were studied intensively, the physiological role of uracil nucleotide-responsive P2Y2, P2Y4, P2Y6, and P2Y14 receptors is still unclear, especially in the cardiovascular system. This study revealed that uridine-5'-triphosphate (UTP) protected cultured rat cardiomyocytes during hypoxia and explored the UTP signaling pathway leading to this cardioprotection. We found that UTP, but not UDP or uridine, significantly reduced cardiomyocyte death induced by hypoxia. Incubation with UTP for 1 h, before exposure to hypoxic conditions, protected the cells 24 h later. The cardioprotective effect of UTP was reduced in the presence of the P2 antagonist suramin. In addition, UTP caused a transient increase of [Ca2+]i in cardiomyocytes. Pyridoxal-5'-phosphate-6-azophenyl-2,4-disulfonate (PPADS) or Reactive blue 2 (RB-2), other antagonists of P2 receptors, abolished the [Ca2+]i elevation caused by UTP. We used various inhibitors of the Ca2+ signaling pathway to show that UTP elevated levels of [Ca2+]i, originating from intracellular sources, via activation of phospholipase C and the IP3 receptor. Interestingly, these inhibitors of the Ca2+ signaling pathway did not prevent the immediate protective effect caused by UTP. Although mitochondrial KATP channels are involved in other preconditioning mediator pathways, the involvement of these channels in the cardioprotective effect induced by UTP was ruled out, because 5-hydroxydecanoic acid (5-HD), a specific inhibitor of these channels, did not prevent the protection.


Assuntos
Cardiotônicos/metabolismo , Hipóxia Celular , Miócitos Cardíacos/metabolismo , Fosfato de Piridoxal/análogos & derivados , Estresse Fisiológico , Nucleotídeos de Uracila/metabolismo , Trifosfato de Adenosina/análise , Trifosfato de Adenosina/metabolismo , Animais , Western Blotting , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática , Imuno-Histoquímica , L-Lactato Desidrogenase/análise , L-Lactato Desidrogenase/metabolismo , Antagonistas do Receptor Purinérgico P2 , Fosfato de Piridoxal/farmacologia , Ratos , Receptores Purinérgicos P2/efeitos dos fármacos , Receptores Purinérgicos P2/metabolismo , Suramina/farmacologia , Fatores de Tempo , Triazinas/farmacologia , Fosfolipases Tipo C/metabolismo , Uridina/metabolismo , Difosfato de Uridina/metabolismo , Uridina Trifosfato/metabolismo
6.
J Pharmacol Exp Ther ; 313(3): 1046-57, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15681657

RESUMO

N,N,N',N'-Tetrakis(2-pyridylmethyl)-ethylenediamine (TPEN), a transition-metal chelator, was recently found to protect against myocardial ischemia-reperfusion injury. The goals of this study were to investigate the in vivo antiarrhythmic and antifibrillatory potential of TPEN in rats and guinea pigs and to study the in vitro effects of TPEN on calcium homeostasis in cultured newborn rat cardiac cells in normoxia and hypoxia. We demonstrated on an in vivo rat model of ischemia-reperfusion that TPEN abolishes ventricular fibrillation incidence and mortality and decreases the incidence and duration of ventricular tachycardia. To elucidate the mechanism of cardioprotection by TPEN, contraction, synchronization, and intracellular calcium level were examined in vitro. We have shown for the first time that TPEN prevented the increase in intracellular Ca(2+) levels ([Ca(2+)](i)) caused by hypoxia and abolished [Ca(2+)](i) elevation caused by high extracellular Ca(2+) levels ([Ca(2+)](o)) or by caffeine. Addition of TPEN returned synchronized beating of cardiomyocytes desynchronized by [Ca(2+)](o) elevation. To discover the mechanism by which TPEN reduces [Ca(2+)](i) in cardiomyocytes, the cells were treated with thapsigargin, which inhibits Ca(2+) uptake into the sarcoplasmic reticulum (SR). TPEN successfully reduced [Ca(2+)](i) elevated by thapsigargin, indicating that TPEN did not sequester Ca(2+) in the SR. However, TPEN did not reduce [Ca(2+)](i) in the Na(+)-free medium in which the Na(+)/Ca(2+) exchanger was inhibited. Taken together, the results show that activation of sarcolemmal Na(+)/Ca(2+) exchanger by TPEN increases Ca(2+) extrusion from the cytoplasm of cardiomyocytes, preventing cytosolic Ca(2+) overload, which explains the beneficial effects of TPEN on postischemic cardiac status.


Assuntos
Cálcio/metabolismo , Quelantes/farmacologia , Etilenodiaminas/farmacologia , Homeostase/efeitos dos fármacos , Isquemia Miocárdica/prevenção & controle , Animais , Antiarrítmicos/farmacologia , Hipóxia Celular , L-Lactato Desidrogenase/metabolismo , Masculino , Mitocôndrias Cardíacas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sódio/metabolismo
7.
Am J Physiol Heart Circ Physiol ; 288(6): H2792-801, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15681707

RESUMO

Activation of either the A(1) or the A(3) adenosine receptor (A(1)R or A(3)R, respectively) elicits delayed cardioprotection against infarction, ischemia, and hypoxia. Mitochondrial contribution to the progression of cardiomyocyte injury is well known; however, the protective effects of adenosine receptor activation in cardiac cells with a respiratory chain deficiency are poorly elucidated. The aim of our study was to further define the role of A(1)R and A(3)R activation on functional tolerance after inhibition of the terminal link of the mitochondrial respiratory chain with sodium azide, in a state of normoxia or hypoxia, compared with the effects of the mitochondrial ATP-sensitive K(+) channel opener diazoxide. Treatment with 10 mM sodium azide for 2 h in normoxia caused a considerable decrease in the total ATP level; however, activation of adenosine receptors significantly attenuated this decrease. Diazoxide (100 muM) was less effective in protection. During treatment of cultured cardiomyocytes with hypoxia in the presence of 1 mM sodium azide, the A(1)R agonist 2-chloro-N(6)-cyclopentyladenosine was ineffective, whereas the A(3)R agonist 2-chloro-N(6)-iodobenzyl-5'-N-methylcarboxamidoadenosine (Cl-IB-MECA) attenuated the decrease in ATP level and prevented cell injury. Cl-IB-MECA delayed the dissipation in the mitochondrial membrane potential during hypoxia in cells impaired in the mitochondrial respiratory chain. In cells with elevated intracellular Ca(2+) concentration after hypoxia and treatment with NaN(3) or after application of high doses of NaN(3), Cl-IB-MECA immediately decreased the elevated intracellular Ca(2+) concentration toward the diastolic control level. The A(1)R agonist was ineffective. This may be especially important for the development of effective pharmacological agents, because mitochondrial dysfunction is a leading factor in the pathophysiological cascade of heart disease.


Assuntos
Coração/fisiologia , Mitocôndrias Cardíacas/fisiologia , Células Musculares/fisiologia , Receptor A1 de Adenosina/fisiologia , Receptor A3 de Adenosina/fisiologia , Agonistas do Receptor A1 de Adenosina , Agonistas do Receptor A3 de Adenosina , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/fisiologia , Hipóxia Celular , Células Cultivadas , Diazóxido/farmacologia , Homeostase , Mitocôndrias Cardíacas/patologia , Células Musculares/citologia , Canais de Potássio/fisiologia , Ratos , Azida Sódica/farmacologia
8.
J Mol Histol ; 35(5): 463-70, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15571324

RESUMO

This study examined whether triiodo-L-thyronine (T3) affects the expression of the major intercellular channel protein, connexin-43, and contractile protein alpha-sarcomeric actin. Cultured cardiomyocytes from newborn rats were treated on day three in culture with 10 or 100 nM T3 and examined 48 and 72 h thereafter. Treated and untreated cells were examined by immunofluorescence and electron microscopy. Expression levels of Cx43 and sarcomeric alpha-actin were monitored by Western blot analysis. Immunofluorescence labeling showed cell membrane location of Cx43 in punctuate gap junctions, whereby fluorescence signal area was significantly higher in cultured cardiomyocytes exposed to T3. This correlated with electron microscopical findings showing increased numbers and size of gap junction profiles, as well as with a significant dose-dependent increase of Cx43 expression detected by Western blot. Immunofluorescence of sarcomeric a-actin was enhanced and its expression increased dose- and time-dependently in T3-treated cultured heart myocytes. However, exposure to the higher dosage (100 nM) of T3 caused mild disintegration of sarcomeric a-actin in some myocytes, suggesting an over-dosage. The results indicate that T3 up-regulates Cx43 and accelerates gap junction formation in cultured neonatal cardiomyocytes. They suggest that thyroid status cannot only modulate the mechanical function of cardiomyocytes but also cell-to-cell communication essential for myocardial electrical and metabolic synchronizations.


Assuntos
Actinas/biossíntese , Conexina 43/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Tri-Iodotironina/farmacologia , Actinas/genética , Animais , Células Cultivadas , Conexina 43/genética , Relação Dose-Resposta a Droga , Miócitos Cardíacos/ultraestrutura , Ratos
9.
Cell Calcium ; 36(5): 387-96, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15451622

RESUMO

Intracellular calcium signaling cascade induced by adenosine A(3) receptor activation was studied in this work. It was found that adenosine A(3) receptor activation (and not A(1) or A(2A) adenosine receptors activation) leads to an increase in cytosolic calcium and its further extrusion. A selective A(3) agonist Cl-IB-MECA (2-chloro-N(6)-(3-iodobenzyl)adenosine-5'-N-methyluronamide) induced an increase in cytoplasmic calcium in a dose-dependent manner, and was independent on extracellular calcium. The Ca(2+) signal in newborn cardiomyocytes, induced by A(3) receptor activation, is dependent on a pertussis toxin-sensitive G-protein. The action of Cl-IB-MECA was not inhibited by an inhibitor of phospholipase C (PLC), and by antagonists to inositol 1,4,5-trisphosphate (IP(3)) receptor. In contrast, inhibition of ryanodine receptor prevented calcium elevation induced by this agonist. It was shown that extrusion of the elevated cytosolic Ca(2+) was achieved via activation of sarcoplasmic reticulum (SR) Ca(2+)-reuptake and of sarcolemmal Na(+)/Ca(2+) exchanger (NCX). The increase in the SR Ca(2+)-uptake and NCX Ca(2+) efflux were sufficient not only for compensation of Ca(2+) release from SR after A(3) receptor activation, but also for an effective prevention of extensive increase in intracellular Ca(2+) and may provide mechanism against cellular Ca(2+) overload. In cells with elevated [Ca(2+)](i) (due to increase of [Ca(2+)](o)), adenosine or Cl-IB-MECA decreased the [Ca(2+)](i) toward diastolic control level, whereas agonist of A(1) receptor was ineffective. The protective effect of A(3) receptor agonist was abolished in the presence of selective A(3) receptor antagonist MRS1523.


Assuntos
Adenosina/análogos & derivados , Sinalização do Cálcio/fisiologia , Miócitos Cardíacos/fisiologia , Receptor A3 de Adenosina/fisiologia , Adenosina/farmacologia , Agonistas do Receptor A3 de Adenosina , Animais , Animais Recém-Nascidos , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Rianodina/farmacologia
10.
Antioxid Redox Signal ; 6(2): 335-44, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15025935

RESUMO

The aim of the present study was to investigate the protective role of pharmacological preconditioning on antioxidant enzymes using A(1) and A(3) adenosine receptor agonists in the recovery of the isolated myocardium after cardioplegic ischemia. Two different modes of preconditioning were studied: isolated rat hearts were perfused with A(1) receptor agonist 2-chloro-N(6)-cyclopentyladenosine (CCPA) or A(3) 2-chloro-N(6)-(3-iodobenzyl) adenosine-5'-N-methyluronamide (Cl-IB-MECA) (1 nM), followed by cardioplegic ischemia and reperfusion (30 min each) (perfusion mode), or CCPA or Cl-IB-MECA (100 micro g/kg) were injected intravenously 24 h before the experiment (injection mode). Hearts treated with CCPA improved in terms of mechanical function, infarct size, ATP levels, superoxide dismutase, and catalase (p < 0.005) in both modes of administration. Cl-IB-MECA was beneficial mainly in the injected group. Reduced damage to the mitochondria in the CCPA-treated hearts was observed using electron microscopy evaluation. In the Cl-IB-MECA-injected hearts, mitochondrial damage was moderate. CCPA in both modes of treatment and Cl-IB-MECA in the injected mode were beneficial in protecting the perfused isolated rat heart, subjected to normothermic cardioplegic ischemia. This protection was partially related to the higher myocardial activity of superoxide dismutase and catalase.


Assuntos
Adenosina/análogos & derivados , Antioxidantes/metabolismo , Precondicionamento Isquêmico Miocárdico , Isquemia Miocárdica/metabolismo , Reperfusão Miocárdica , Miocárdio/metabolismo , Receptor A1 de Adenosina/metabolismo , Receptor A3 de Adenosina/metabolismo , Adenosina/análise , Adenosina/farmacologia , Agonistas do Receptor A1 de Adenosina , Agonistas do Receptor A3 de Adenosina , Trifosfato de Adenosina/metabolismo , Animais , Catalase/metabolismo , Glutationa Peroxidase/metabolismo , Coração/efeitos dos fármacos , Hemodinâmica , Técnicas In Vitro , Masculino , Miocárdio/patologia , Miocárdio/ultraestrutura , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo
11.
Mol Cell Biochem ; 254(1-2): 311-8, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14674711

RESUMO

Elevated glycogen levels in heart have been shown to have cardioprotective effects against ischemic injury. We have therefore established a model for elevating glycogen content in primary rat cardiac cells grown in culture and examined potential mechanisms for the elevation (glycogen supercompensation). Glycogen was depleted by exposing the cells to hypoxia for 2 h in the absence of glucose in the medium. This was followed by incubating the cells with 28 mM glucose in normoxia for up to 120 h. Hypoxia decreased glycogen content to about 15% of control, oxygenated cells. This was followed by a continuous increase in glycogen in the hypoxia treated cells during the 120 h recovery period in normoxia. By 48 h after termination of hypoxia, the glycogen content had returned to baseline levels and by 120 h glycogen was about 150% of control. The increase in glycogen at 120 h was associated with comparable relative increases in glucose uptake (approximately 180% of control) and the protein level of the glut-1 transporter (approximately 170% of control), whereas the protein level of the glut-4 transporter was decreased to < 10% of control. By 120 h, the hypoxia-treated cells also exhibited marked increases in the total (approximately 170% of control) and fractional activity of glycogen synthase (control, approximately 15%; hypoxia-treated, approximately 30%). Concomitantly, the hypoxia-treated cells also exhibited marked decreases in the total (approximately 50% of control) and fractional activity of glycogen phosphorylase (control, approximately 50%; hypoxia-treated, approximately 25%). Thus, we have established a model of glycogen supercompensation in cultures of cardiac cells that is explained by concerted increases in glucose uptake and glycogen synthase activity and decreases in phosphorylase activity. This model should prove useful in studying the cardioprotective effects of glycogen.


Assuntos
Glicogênio/metabolismo , Hipóxia , Proteínas Musculares , Miocárdio/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Creatina Quinase/metabolismo , Glucose/metabolismo , Transportador de Glucose Tipo 1 , Transportador de Glucose Tipo 4 , Glicogênio Fosforilase/metabolismo , Glicogênio Sintase/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Ratos , Fatores de Tempo
12.
Mol Cell Biochem ; 252(1-2): 133-9, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14577586

RESUMO

Adenosine has been found to be cardioprotective during episodes of cardiac ischemia/reperfusion through activation of the A1 and possibly A1 receptors. Therefore, we have investigated whether activation of these receptors can protect also against apoptotic death induced by angiotensin II (Ang II) in neonatal rat cardiomyocyte cultures. Exposure to Ang II (10 nM) resulted in a 3-fold increase in programmed cell death (p < 0.05). Pretreatment with the A1 adenosine receptor agonist 2-chloro-N6-cyclopentyladenosine (CCPA, 1 microM), abolished the effects of Ang II on programmed cardiomyocyte death. Moreover, exposure of cells to the A1 adenosine receptor antagonist 8-cyclopentyl- 1,3-dipropylxanthine (CPX) before pretreatment with CCPA, prevented the protective effect of the latter. Pretreatment with the A3 adenosine receptor agonist N6-(3-iodobenzyl) adenosine-5'-N-methyluronamide (IB-MECA, 0.1 microM), led to a partial decrease in apoptotic rate induced by Ang II. Exposure of myocytes to Ang II caused an immediate increase in the concentration of intracellular free Ca2+ that lasted 40-60 sec. Pretreatment of cells with CCPA or IB-MECA did not block Ang II-induced Ca2+ elevation. In conclusion, activation of adenosine A1 receptors can protect the cardiac cells from apoptosis induced by Ang II, while activation of the adenosine A3 receptors confers partial cardioprotection.


Assuntos
Adenosina/farmacologia , Angiotensina II/farmacologia , Apoptose/efeitos dos fármacos , Coração/efeitos dos fármacos , Miocárdio/citologia , Agonistas do Receptor A1 de Adenosina , Agonistas do Receptor A3 de Adenosina , Animais , Cálcio/metabolismo , Células Cultivadas , Marcação In Situ das Extremidades Cortadas , Miocárdio/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A3 de Adenosina/efeitos dos fármacos
13.
Mol Cell Biochem ; 250(1-2): 11-9, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12962138

RESUMO

A model to study glycogen supercompensation (the significant increase in glycogen content above basal level) in primary rat skeletal muscle culture was established. Glycogen was completely depleted in differentiated myotubes by 2 h of electrical stimulation or exposure to hypoxia during incubation in medium devoid of glucose. Thereafter, cells were incubated in medium containing glucose, and glycogen supercompensation was clearly observed in treated myotubes after 72 h. Peak glycogen levels were obtained after 120 h, averaging 2.5 and 4 fold above control values in the stimulated- and hypoxia-treated cells, respectively. Glycogen synthase activity increased and phosphorylase activity decreased continuously during 120 h of recovery in the treated cells. Rates of 2-deoxyglucose uptake were significantly elevated in the treated cells at 96 and 120 h, averaging 1.4-2 fold above control values. Glycogenin content increased slightly in the treated cells after 48 h (1.2 fold vs. control) and then increased considerably, achieving peak values after 120 h (2 fold vs. control). The results demonstrate two phases of glycogen supercompensation: the first phase depends primarily on activation of glycogen synthase and inactivation of phosphorylase; the second phase includes increases in glucose uptake and glycogenin level.


Assuntos
Glicogênio/química , Músculo Esquelético/citologia , Trifosfato de Adenosina/metabolismo , Animais , Diferenciação Celular , Desoxiglucose/farmacocinética , Estimulação Elétrica , Glucose/metabolismo , Glucose/farmacocinética , Glicogênio/metabolismo , Glicogênio Fosforilase/metabolismo , Glicogênio Sintase/metabolismo , Hipóxia , Ratos , Fatores de Tempo
14.
J Biol Chem ; 278(42): 40917-22, 2003 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-12851407

RESUMO

Low energy visible light (LEVL) irradiation has been shown to exert some beneficial effects on various cell cultures. For example, it increases the fertilizing capability of sperm cells, promotes cell proliferation, induces sprouting of neurons, and more. To learn about the mechanism of photobiostimulation, we studied the relationship between increased intracellular calcium ([Ca2+]i) and reactive oxygen species production following LEVL illumination of cardiomyocytes. We found that visible light causes the production of O2. and H2O2 and that exogenously added H2O2 (12 microm) can mimic the effect of LEVL (3.6 J/cm2) to induce a slow and transient increase in [Ca2+]i. This [Ca2+]i elevation can be reduced by verapamil, a voltage-dependent calcium channel inhibitor. The kinetics of [Ca2+]i elevation and morphologic damage following light or addition of H2O2 were found to be dose-dependent. For example, LEVL, 3.6 J/cm2, which induced a transient increase in [Ca2+]i, did not cause any cell damage, whereas visible light at 12 J/cm2 induced a linear increase in [Ca2+]i and damaged the cells. The linear increase in [Ca2+]i resulting from high energy doses of light could be attenuated into a non-linear small rise in [Ca2+]i by the presence of extracellular catalase during illumination. We suggest that the different kinetics of [Ca2+]i elevation following various light irradiation or H2O2 treatment represents correspondingly different adaptation levels to oxidative stress. The adaptive response of the cells to LEVL represented by the transient increase in [Ca2+]i can explain LEVL beneficial effects.


Assuntos
Cálcio/metabolismo , Miocárdio/citologia , Espécies Reativas de Oxigênio , Animais , Divisão Celular , Sobrevivência Celular , Células Cultivadas , Desmina/química , Relação Dose-Resposta à Radiação , Espectroscopia de Ressonância de Spin Eletrônica , Peróxido de Hidrogênio/metabolismo , Cinética , L-Lactato Desidrogenase/metabolismo , Luz , Magnetismo , Miocárdio/metabolismo , Oxigênio/metabolismo , Ratos , Fatores de Tempo
15.
J Mol Cell Cardiol ; 34(5): 493-507, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12056854

RESUMO

Recently, we reported that the activation of A(3) adenosine receptor (A(3)R) in newborn cultured cardiomyocytes by highly selective agonist Cl-IB-MECA (2-chloro-N(6)-(3-iodobenzyl)adenosine-5'-N-methyluronamide) induces protection against the anthracycline antibiotic doxorubicin (DOX) cardiotoxicity. The present study was undertaken to further characterize the cardioprotective action of A(3)R activation by revealing the structural changes in cardiomyocytes elicited upon exposure to DOX. Morphological observations (ultrastructural and immunocytochemical) indicate that after DOX treatment, the cardiomyocytes undergo destructive alterations, and protective action of A(3)R is not connected with its anti-apoptotic activity. A(3)R activation appeared to prevent destructive alterations of cardiomyocyte mitochondria and dissipation of mitochondrial membrane potential. In DOX-treated cardiomyocytes, appearance of disorganized desmin and contractile filaments was related to detrimental alterations in the mitochondrial structure, in particular their position and transmembrane potential. In intact cardiomyocytes, diazoxide, a selective mitochondrial K(ATP) channel opener, induced an increase in ATP synthesis within 15 min of application. Similar effect was obtained by activation of adenosine A(1)R. However, A(3)R agonist Cl-IB-MECA did not affect ATP synthesis. Neither A(1)R agonist CCPA (2-chloro-N(6)-cyclopentyladenosine) nor diazoxide protected cardiomyocytes from the detrimental effects of DOX. Thus, the opening of mitochondrial K(ATP) channels does not seem to be effective during the slow development of anthracycline cytotoxicity. Our results indicate that DOX increases the activity of lysosomes, which may contribute to cell injury in an "oncotic" manner and also demonstrate the proinflammatory potency of the drug. Furthermore, the decreased acidification of cytoplasm upon activation of A(3)R may attenuate the ongoing inflammatory response. The present study identifies a novel role for A(3)R selective agonist Cl-IB-MECA and suggests its importance in regulating cardiac cellular function.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Miocárdio/metabolismo , Receptores Purinérgicos P1/metabolismo , Animais , Antibióticos Antineoplásicos/toxicidade , Morte Celular , Células Cultivadas , Fragmentação do DNA , Doxorrubicina/toxicidade , Resistência a Medicamentos/fisiologia , Marcação In Situ das Extremidades Cortadas , Microscopia de Fluorescência , Mitocôndrias/metabolismo , Miocárdio/patologia , Miocárdio/ultraestrutura , Agonistas do Receptor Purinérgico P1 , Ratos
16.
Exp Clin Cardiol ; 7(2-3): 138-45, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-19649238

RESUMO

OBJECTIVES: Cardioprotection (delaying of irreversible damage in hypoxia or prevention of doxorubicin [DOX] toxicity) is achieved by increasing the energy supply, or decreasing the energy demand in the cell and may be regulated through adenosine (ADO) receptor (AR) signalling. The aim of this study was to define of the protective role of ADO A(1)R and A(3)R against these two different kinds of stress conditions via direct action on isolated cardiomyocytes. Effects of A(1) and A(3) adenosine receptors were assessed by comparing morphological-functional tolerance, cellular energy state and contribution of the mitochondrial K(ATP) channels during development of hypoxia and DOX cytotoxicity. METHODS: The primary cardiac myocyte cultures were treated in a hypoxic chamber of N(2) (100%) in glucose-free media. A second group of cells were treated on day 4 in culture with 0.5 to 5 muM DOX for 18 h and then incubated in drug-free growth medium for an additional 24 h or 72 h. The hypoxic and cytotoxic damage was characterized by morphological and biochemical evaluations. RESULTS: The A(1)R and A(3)R selective agonists (CCPA and Cl-IB-MECA, respectively) significantly decreased damage to cardiac myocytes under hypoxic conditions. Activation of both A(1)R and A(3)R together (100 nM) was more efficient in protection against hypoxia than by each one alone. The A(3)R agonist Cl-IB-MECA (100 nM) shows cardioprotective activity to the DOX-treated cells; however, the A(1)R agonist CCPA (10 nM to 10 muM) was not effective in protection against DOX toxicity. CONCLUSION: Activation of both the ADO receptors (A(1)R and A(3)R) leads to positive beneficial effects in cultured cardiomyocytes in 90 min hypoxia, but only A(3)R activation renders positive response against slowly developed DOX toxicity. Hence, the cascade of events involved in cardioprotection appears to be distinct for A(1) and A(3) receptor signalling.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...