Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biol Pharm Bull ; 41(11): 1685-1693, 2018 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-30197410

RESUMO

Echinacoside is a major compound of Cistanche Herb and has glutamate release-inhibiting activity in the brain. Given the involvement of excitotoxicity caused by massive glutamate in the pathophysiology of epilepsy, we explored the antiepileptic effect of echinacoside on kainic acid-induced seizures in rats. The rats were intraperitoneally administrated echinacoside for 30 min prior to intraperitoneal injection with kainic acid. The results showed that kainic acid induced seizure-like behavioral patterns, increased glutamate concentrations, caused neuronal loss and microglial activation, and stimulated proinflammatory cytokine gene expression in the hippocampus. These kainic acid-induced alternations were found to be attenuated by echinacoside pretreatment. Furthermore, decreased Akt and glycogen synthase kinase 3ß (GSK3ß) phosphorylation as well as Bcl-2 expression in the hippocampus was reversed by the echinacoside pretreatment. These results demonstrate that echinacoside exert its antiepileptic and neuroprotective actions in a kainic acid rat model through suppressing inflammatory response and activating the Akt/GSK3ß signaling. Therefore, the present study suggests that echinacoside is the potentially useful in the prevention of epilepsy.


Assuntos
Encéfalo/efeitos dos fármacos , Cistanche/química , Epilepsia , Glicogênio Sintase Quinase 3 beta/metabolismo , Glicosídeos/farmacologia , Inflamação/prevenção & controle , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Epilepsia/etiologia , Epilepsia/metabolismo , Epilepsia/patologia , Epilepsia/prevenção & controle , Ácido Glutâmico/efeitos adversos , Ácido Glutâmico/metabolismo , Glicosídeos/uso terapêutico , Inflamação/induzido quimicamente , Inflamação/metabolismo , Ácido Caínico , Masculino , Microglia/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/patologia , Síndromes Neurotóxicas/prevenção & controle , Fosforilação , Fitoterapia , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Convulsões/metabolismo , Convulsões/prevenção & controle , Transdução de Sinais
2.
Artigo em Inglês | MEDLINE | ID: mdl-29803926

RESUMO

CGS12066, a 5-hydroxytryptamine 1B (5-HT1B) receptor agonist, has been reported to exhibit antidepressant activity. Considering that glutamatergic dysfunction is implicated in depression, the effect of CGS12066 on glutamate release in rat hippocampal nerve terminals and possible underlying mechanism were investigated. We observed that CGS12066 inhibited 4-aminopyridine (4-AP)-evoked glutamate release, and that a 5-HT1B receptor antagonist blocked this inhibition. Western blot analysis and immunocytochemistry confirmed the presence of presynaptic 5-HT1B receptor proteins. CGS12066-mediated inhibition of 4-AP-evoked glutamate release was completely abolished in the synaptosomes pretreated with inhibitors of Gi/Go-protein, adenylate cyclase (AC), and protein kinase A (PKA), namely pertussis toxin, MDL12330A, and H89, respectively. CGS12066 reduced the elevation of 4-AP-evoked intrasynaptosomal Ca2+ and cyclic AMP (cAMP) levels, but did not affect the synaptosomal membrane potential. Furthermore, in the presence of ω-conotoxin MVIIC, a N- and P/Q-type channel blocker, CGS12066-mediated inhibition of 4-AP-evoked glutamate release was markedly reduced; however, the intracellular Ca2+-release inhibitors dantrolene and CGP37157 did not affect the CGS12066 effect. Furthermore, CGS12066 reduced glutamatergic miniature excitatory postsynaptic current (mEPSC) frequency but did not affect mEPSC amplitude or glutamate-activated currents in hippocampal slices. Our data are the first to suggest that CGS12066 reduces AC/cAMP/PKA activation, through the activation of Gi/Go protein-coupled 5-HT1B receptors present on hippocampal nerve terminals, subsequently reducing Ca2+ entry through voltage-dependent Ca2+ channels and reducing 4-AP-evoked glutamate release. This investigation into the role of 5-HT1B receptors in glutamate release provides crucial information regarding the potential therapeutic role of 5-HT1B receptors for treating depression.


Assuntos
Ácido Glutâmico/metabolismo , Hipocampo/efeitos dos fármacos , Quinoxalinas/farmacologia , Agonistas do Receptor 5-HT1 de Serotonina/farmacologia , Sinapses/efeitos dos fármacos , Sinaptossomos/efeitos dos fármacos , Adenilil Ciclases/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Hipocampo/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Ratos Sprague-Dawley , Receptor 5-HT1B de Serotonina/metabolismo , Sinapses/metabolismo , Sinaptossomos/metabolismo , Técnicas de Cultura de Tecidos
3.
Food Funct ; 7(1): 212-26, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26667007

RESUMO

This study examined whether xanthohumol, a hop-derived prenylated flavonoid present in beer, affects glutamate release in the rat hippocampus. In the rat hippocampal nerve terminals (synaptosomes), xanthohumol inhibited the release of 4-aminopyridine (4-AP)-evoked glutamate and the elevation of cytosolic Ca(2+) concentration, whereas it had no effect on 4-AP-mediated depolarization. The inhibitory effect of xanthohumol on the evoked glutamate release was prevented by removing extracellular Ca(2+), using the Cav2.2 (N-type) and Cav2.1 (P/Q-type) channel blocker ω-CgTX MVIIC, the calmodulin antagonists W7 and calmidazolium, and the protein kinase A inhibitor H89; however, no such effect was observed when the G-protein inhibitor N-ethylmaleimide was used. In addition, immunocytochemical data demonstrated that GABAA receptors are present in the hippocampal synaptosomes and that the xanthohumol effect on evoked glutamate release was antagonized by the GABAA receptor antagonist SR95531. Furthermore, in slice preparations, xanthohumol reduced the frequency of miniature excitatory postsynaptic currents without affecting their amplitude. We conclude that xanthohumol acts at GABAA receptors present in the hippocampal nerve terminals to decrease the Ca(2+) influx through N- and P/Q-type Ca(2+) channels, which subsequently suppresses the Ca(2+)-calmodulin/PKA cascade to decrease the evoked glutamate release.


Assuntos
Flavonoides/farmacologia , Ácido Glutâmico/metabolismo , Hipocampo/ultraestrutura , Terminações Pré-Sinápticas/efeitos dos fármacos , Propiofenonas/farmacologia , 4-Aminopiridina/farmacologia , Animais , Cerveja/análise , Cálcio/análise , Cálcio/fisiologia , Bloqueadores dos Canais de Cálcio/farmacologia , Calmodulina/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Antagonistas de Receptores de GABA-A/farmacologia , Hipocampo/metabolismo , Imidazóis/farmacologia , Masculino , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/fisiologia , Sinaptossomos/química , Sinaptossomos/fisiologia
4.
Int J Mol Sci ; 16(3): 5555-71, 2015 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-25768340

RESUMO

The effect of palmitoylethanolamide (PEA), an endogenous fatty acid amide displaying neuroprotective actions, on glutamate release from rat cerebrocortical nerve terminals (synaptosomes) was investigated. PEA inhibited the Ca²âº-dependent release of glutamate, which was triggered by exposing synaptosomes to the potassium channel blocker 4-aminopyridine. This release inhibition was concentration dependent, associated with a reduction in cytosolic Ca²âº concentration, and not due to a change in synaptosomal membrane potential. The glutamate release-inhibiting effect of PEA was prevented by the Ca(v)2.1 (P/Q-type) channel blocker ω-agatoxin IVA or the protein kinase A inhibitor H89, not affected by the intracellular Ca²âº release inhibitors dantrolene and CGP37157, and partially antagonized by the cannabinoid CB1 receptor antagonist AM281. Based on these results, we suggest that PEA exerts its presynaptic inhibition, likely through a reduction in the Ca²âº influx mediated by Ca(v)2.1 (P/Q-type) channels, thereby inhibiting the release of glutamate from rat cortical nerve terminals. This release inhibition might be linked to the activation of presynaptic cannabinoid CB1 receptors and the suppression of the protein kinase A pathway.


Assuntos
Analgésicos/farmacologia , Agonistas de Receptores de Canabinoides/farmacologia , Etanolaminas/farmacologia , Ácido Glutâmico/metabolismo , Ácidos Palmíticos/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Tratos Piramidais/efeitos dos fármacos , Amidas , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Antagonistas de Receptores de Canabinoides/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Exocitose , Masculino , Bloqueadores dos Canais de Potássio/farmacologia , Terminações Pré-Sinápticas/metabolismo , Tratos Piramidais/metabolismo , Ratos , Ratos Sprague-Dawley
5.
Eur J Pharmacol ; 755: 6-15, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25746462

RESUMO

Hispidulin is a flavonoid compound which is an active ingredient in a number of traditional Chinese medicinal herbs, and it has been reported to inhibit glutamate release. The purpose of this study was to investigate whether hispidulin protects against seizures induced by kainic acid, a glutamate analog with excitotoxic properties. The results indicated that intraperitoneally administering hispidulin (10 or 50mg/kg) to rats 30 min before intraperitoneally injecting kainic acid (15 mg/kg) increased seizure latency and decreased seizure score. In addition, hispidulin substantially attenuated kainic acid-induced hippocampal neuronal cell death, and this protective effect was accompanied by the suppression of microglial activation and the production of proinflammatory cytokines such as interleukin-1ß, interleukin-6, and tumor necrosis factor-α in the hippocampus. Moreover, hispidulin reduced kainic acid-induced c-Fos expression and the activation of mitogen-activated protein kinases in the hippocampus. These data suggest that hispidulin has considerable antiepileptic, neuroprotective, and antiinflammatory effects on kainic acid-induced seizures in rats.


Assuntos
Anti-Inflamatórios/uso terapêutico , Anticonvulsivantes/uso terapêutico , Flavonas/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Convulsões/tratamento farmacológico , Animais , Anti-Inflamatórios/farmacologia , Anticonvulsivantes/farmacologia , Região CA3 Hipocampal/efeitos dos fármacos , Região CA3 Hipocampal/metabolismo , Região CA3 Hipocampal/patologia , Morte Celular/efeitos dos fármacos , Citocinas/genética , Flavonas/farmacologia , Ácido Caínico , Masculino , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Convulsões/metabolismo , Convulsões/patologia
6.
J Pharmacol Exp Ther ; 351(1): 134-45, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25047516

RESUMO

The excitotoxicity caused by excessive glutamate is a critical element in the neuropathology of acute and chronic brain disorders. Therefore, inhibition of glutamate release is a potentially valuable therapeutic strategy for treating these diseases. In this study, we investigated the effect of celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor that reduces the level of prostaglandin E2 (PGE2), on endogenous glutamate release in rat cerebral cortex nerve terminals (synaptosomes). Celecoxib substantially inhibited the release of glutamate induced by the K(+) channel blocker 4-aminopyridine (4-AP), and this phenomenon was prevented by chelating the extracellular Ca(2+) ions and by the vesicular transporter inhibitor bafilomycin A1. Celecoxib inhibited a 4-AP-induced increase in cytosolic-free Ca(2+) concentration, and the celecoxib-mediated inhibition of glutamate release was prevented by the Cav2.2 (N-type) and Cav2.1 (P/Q-type) channel blocker ω-conotoxin MVIIC. However, celecoxib did not alter 4-AP-mediated depolarization and Na(+) influx. In addition, this glutamate release-inhibiting effect of celecoxib was mediated through the PGE2 subtype 2 receptor (EP2) because it was not observed in the presence of butaprost (an EP2 agonist) or PF04418948 [1-(4-fluorobenzoyl)-3-[[6-methoxy-2-naphthalenyl)methyl]-3-azetidinecarboxylic acid; an EP2 antagonist]. The celecoxib effect on 4-AP-induced glutamate release was prevented by the inhibition or activation of protein kinase A (PKA), and celecoxib decreased the 4-AP-induced phosphorylation of PKA. We also determined that COX-2 and the EP2 receptor are present in presynaptic terminals because they are colocalized with synaptophysin, a presynaptic marker. These results collectively indicate that celecoxib inhibits glutamate release from nerve terminals by reducing voltage-dependent Ca(2+) entry through a signaling cascade involving EP2 and PKA.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Ácido Glutâmico/metabolismo , Terminações Pré-Sinápticas/efeitos dos fármacos , Prostaglandinas E/metabolismo , Pirazóis/farmacologia , Receptores de Prostaglandina E/metabolismo , Sulfonamidas/farmacologia , Animais , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/metabolismo , Celecoxib , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Exocitose , Masculino , Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Terminações Pré-Sinápticas/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inibidores , Sódio/metabolismo , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo
7.
J Ethnopharmacol ; 147(2): 488-96, 2013 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-23542145

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Danshen is a commonly used traditional Chinese medicine and has received considerable attention due to their beneficial effects on the health, including prevention of cardiovascular disease, and cancer. Tanshinone IIA, a major active constituent of Danshen, has been reported to have a neuroprotective profile. AIM OF THE STUDY: An excessive release of glutamate is considered to be related to neuropathology of several neurological diseases. In this study, we investigated whether tanshinone IIA could affect endogenous glutamate release and explored the possible mechanism. MATERIALS AND METHODS: The experimental model was the isolated nerve terminals (synaptosomes) purified from the rat cerebral cortex. The release of glutamate was evoked by the K(+) channel blocker 4-aminopyridine (4-AP) and measured by one-line enzyme-coupled fluorometric assay. We also used a membrane potential-sensitive dye to assay nerve terminal excitability and depolarization, and a Ca(2+) indicator, Fura-2-acetoxymethyl ester, to monitor cytosolic Ca(2+) concentrations ([Ca(2+)]C). RESULTS: Tanshinone IIA inhibited the release of glutamate evoked by 4-AP in a concentration-dependent manner. Inhibition of glutamate release by tanshinone IIA was prevented by the chelating the extracellular Ca(2+) ions, and by the vesicular transporter inhibitor bafilomycin A1. However, the glutamate transporter inhibitor DL-threo-beta-benzyl-oxyaspartate did not have any effect on the action of tanshinone IIA. Tanshinone IIA decreased the depolarization-induced increase in [Ca(2+)]C, whereas it did not alter the resting synaptosomal membrane potential or 4-AP-mediated depolarization. Furthermore, the effect of tanshinone IIA on evoked glutamate release was prevented by the Cav2.2 (N-type) and Cav2.1 (P/Q-type) channel blocker ω-conotoxin MVIIC, but not by the ryanodine receptor blocker dantrolene or the mitochondrial Na(+)/Ca(2+) exchanger blocker CGP37157. Mitogen-activated protein kinase (MEK) inhibition also prevented the inhibitory effect of tanshinone IIA on evoked glutamate release. CONCLUSION: These results show that tanshinone IIA inhibits glutamate release from cortical synaptosomes in rats through the suppression of presynaptic voltage-dependent Ca(2+) entry and MEK signaling cascade.


Assuntos
Abietanos/farmacologia , Córtex Cerebral/citologia , Ácido Glutâmico/metabolismo , Sinaptossomos/metabolismo , 4-Aminopiridina/farmacologia , Animais , Cálcio/metabolismo , Membrana Celular/fisiologia , Medicamentos de Ervas Chinesas , Masculino , Potenciais da Membrana , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Salvia miltiorrhiza
8.
Int J Mol Sci ; 13(7): 9097-9109, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22942754

RESUMO

Curcumin, one of the major constituents of Curcuma longa, has been shown to inhibit depolarization-evoked glutamate release from rat prefrontocortical nerve terminals by reducing voltage-dependent Ca(2+) entry. This study showed that curcumin inhibited ionomycin-induced glutamate release and KCl-evoked FM1-43 release, suggesting that some steps after Ca(2+) entry are regulated by curcumin. Furthermore, disrupting the cytoskeleton organization using cytochalasin D abolished the inhibitory action of curcumin on ionomycin-induced glutamate release. Mitogen-activated protein kinase kinase (MEK) inhibition also prevented the inhibitory effect of curcumin on ionomycin-induced glutamate release. Western blot analyses showed that curcumin decreased the ionomycin-induced phosphorylation of extracellular signal-regulated kinase 1 and 2 (ERK1/2) and synaptic vesicle-associated protein synapsin I, the main presynaptic target of ERK. These results show that curcumin-mediated inhibition of glutamate release involves modulating downstream events by controlling synaptic vesicle recruitment and exocytosis, possibly through a decrease of MAPK/ERK activation and synapsin I phosphorylation, thereby decreasing synaptic vesicle availability for exocytosis.


Assuntos
Antineoplásicos/farmacologia , Curcumina/farmacologia , Exocitose/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Nervos Periféricos/metabolismo , Vesículas Sinápticas/metabolismo , Animais , Cálcio/metabolismo , Ativação Enzimática , Masculino , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sinapsinas/metabolismo
9.
Neurochem Int ; 60(2): 105-14, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22142530

RESUMO

This study was aimed at examining the effect of tamoxifen, a selective estrogen receptor modulator, on the release of endogenous glutamate in rat cerebral cortex nerve terminals (synaptosomes) and exploring the possible mechanism. Tamoxifen inhibited the release of glutamate that was evoked by the K(+) channel blocker 4-aminopyridine (4-AP), and this phenomenon was concentration-dependent and insensitive to the estrogen receptor antagonist. The effect of tamoxifen on the evoked glutamate release was prevented by the chelating extracellular Ca(2+) ions, and by the vesicular transporter inhibitor bafilomycin A1. However, the glutamate transporter inhibitor dl-threo-beta-benzyloxyaspartate did not have any effect on the action of tamoxifen. Tamoxifen did not alter the resting synaptosomal membrane potential or 4-AP-mediated depolarization whereas it decreased the 4-AP-induced increase in cytosolic [Ca(2+)]. Furthermore, the inhibitory effect of tamoxifen on the evoked glutamate release was abolished by the Ca(v)2.2 (N-type) and Ca(v)2.1 (P/Q-type) channel blocker ω-conotoxin MVIIC, but not by the ryanodine receptor blocker dantrolene, or the mitochondrial Na(+)/Ca(2+) exchanger blocker CGP37157. In addition, the protein kinase C (PKC) inhibitors GF109203X or Ro318220 prevented tamoxifen from inhibiting glutamate release. Western blotting showed that tamoxifen significantly decreased the 4-AP-induced phosphorylation of PKC and PKCα. Together, these results suggest that tamoxifen inhibits glutamate release from rat cortical synaptosomes, through the suppression of presynaptic voltage-dependent Ca(2+) entry and PKC activity.


Assuntos
Canais de Cálcio/metabolismo , Córtex Cerebral/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Inibição Neural/efeitos dos fármacos , Terminações Pré-Sinápticas/efeitos dos fármacos , Proteína Quinase C-alfa/antagonistas & inibidores , Tamoxifeno/farmacologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/fisiologia , Córtex Cerebral/enzimologia , Córtex Cerebral/metabolismo , Ácido Glutâmico/fisiologia , Masculino , Inibição Neural/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Terminações Pré-Sinápticas/enzimologia , Terminações Pré-Sinápticas/metabolismo , Proteína Quinase C-alfa/fisiologia , Ratos , Ratos Sprague-Dawley
10.
Chin J Physiol ; 51(5): 308-16, 2008 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-19175187

RESUMO

It is unknown if short-term and long-term intermittent hypobaric hypoxic challenges both exert pro-apoptotic effects on Fas death receptor-dependent apoptotic pathway in rat hearts. Seventy-two Sprague-Dawley rats were randomly assigned into two groups. First, short-term intermittent hypobaric hypoxia (STIHH)-normobaric normoxia (n = 12), hypobaric hypoxia (380 mmHg, 12% O2, 8 hrs/day) for 1 day (n = 12), and for 4 days (n = 12) and second, long-term intermittent hypobaric hypoxia (LTIHH)-normobaric normoxia (n = 12), hypobaric hypoxia for 1 week (n = 12) and 2 weeks (n = 12). After STIHH or LTIHH challenge, Fas receptor related pathway and histopathological analysis in the excised left ventricle was determined by Western blotting, RT-PCR, Hematoxylin-eosin staining, Masson trichrome staining and TUNEL assay. Fas death receptor and TNFalpha were significantly decreased after STIHH whereas Fas receptor, TNFalpha, FAS-associated death domain (FADD), and caspase 8 were increased after LTIHH. In addition, cardiomyocyte disarray and fibrosis were observed in 1 week LTIHH. Cardiac hypertrophy and more severe disarray, fibrosis and cardiac apoptotic activities were observed in 2 week LTIHH. STIHH exerts anti-apoptotic effects on hearts such as downregulation of TNFalpha and Fas receptor whereas LTIHH exerts pro-apoptotic effects such as upregulation of TNFalpha and Fas-mediated apoptotic pathways and lead to cardiac fibrosis and apoptosis. Our findings imply that short-term versus long-term intermittent hypobaric hypoxia exerted protective versus deleterious effects on hearts.


Assuntos
Apoptose , Hipóxia/patologia , Receptor fas/fisiologia , Animais , Cardiomiopatias/patologia , Proteína Ligante Fas/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Fibrose , Coração/fisiologia , Hipóxia/fisiopatologia , Masculino , Miocárdio/patologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Fator de Necrose Tumoral alfa/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA