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










Base de dados
Intervalo de ano de publicação
1.
Neuropharmacology ; 110(Pt A): 308-321, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27515806

RESUMO

Anxiety is an affective disorder that is commonly observed after irreversible brain damage induced by cerebral ischemia and can delay the physical and cognitive recovery, which affects the quality of life of both the patient and family members. However, anxiety after ischemia has received less attention, and mechanisms underlying anxiety-like behaviours induced by chronic cerebral ischemia are under-investigated. In the present study, the chronic cerebral hypoperfusion model was established by the permanent occlusion of the bilateral common carotid arteries (two-vessel occlusion, 2VO) in rats, and anxiety-related behaviours were evaluated. Results indicated that 2VO induced obvious anxiety-like behaviours; the surface expressions of GABAB2 subunits were down-regulated; Brain derived neurotrophic factor (BDNF), tyrosine kinase B (TrkB) and neural cell adhesion molecule (NCAM) were reduced; Meanwhile, the surface expressions of G protein-activated inwardly rectifying potassium (GIRK, Kir3) channels were up-regulated in hippocampal CA1 in 2VO rats. Baclofen, a GABAB receptor agonist, significantly ameliorated the anxiety-like behaviours. It also improved the down-regulation of GABAB2 surface expressions, restored the levels of BDNF, TrkB and NCAM, and reversed the increased surface expressions of Kir3 in hippocampal CA1 in 2VO rats. However, the effects of baclofen were absent in shRNA-GABAB2 infected 2VO rats. These results suggested that activation of GABAB2 subunits could improve BDNF signalling and reverse Kir3 channel surface expressions in hippocampal CA1, which may alleviate the anxiety-like behaviours in rats with chronic cerebral hypoperfusion.


Assuntos
Ansiedade/metabolismo , Doenças das Artérias Carótidas/metabolismo , Receptores de GABA-B/metabolismo , Animais , Ansiedade/tratamento farmacológico , Ansiedade/etiologia , Baclofeno/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/metabolismo , Doenças das Artérias Carótidas/tratamento farmacológico , Doenças das Artérias Carótidas/psicologia , Artéria Carótida Primitiva , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Agonistas dos Receptores de GABA-B/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Proteínas do Tecido Nervoso/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Psicotrópicos/farmacologia , Ratos Sprague-Dawley , Receptor trkB/metabolismo , Receptores de GABA-B/genética , Proteína da Zônula de Oclusão-2/metabolismo
2.
Neural Regen Res ; 11(5): 779-86, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27335562

RESUMO

The selective hyperpolarization-activated cyclic nucleotide-gated (HCN) channel blocker 4-(N-ethyl-N-phenylamino)-1,2-dimethyl-6-(methylamino) pyrimidinium chloride (ZD7288) blocks the induction of long-term potentiation in the perforant path-CA3 region in rat hippocampus in vivo. To explore the mechanisms underlying the action of ZD7288, we recorded excitatory postsynaptic potentials in perforant path-CA3 synapses in male Sprague-Dawley rats. We measured glutamate content in the hippocampus and in cultured hippocampal neurons using high performance liquid chromatography, and determined intracellular Ca(2+) concentration [Ca(2+)]i) using Fura-2. ZD7288 inhibited the induction and maintenance of long-term potentiation, and these effects were mirrored by the nonspecific HCN channel blocker cesium. ZD7288 also decreased glutamate release in hippocampal tissue and in cultured hippocampal neurons. Furthermore, ZD7288 attenuated glutamate-induced rises in [Ca(2+)]i in a concentration-dependent manner and reversed 8-Br-cAMP-mediated facilitation of these glutamate-induced [Ca(2+)]i rises. Our results suggest that ZD7288 inhibits hippocampal synaptic plasticity both glutamate release and resultant [Ca(2+)]i increases in rat hippocampal neurons.

3.
CNS Neurosci Ther ; 21(11): 905-13, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26494128

RESUMO

AIMS: Brain ischemia activates astrocytes in a process known as astrogliosis. Although this process has beneficial effects, excessive astrogliosis can impair neuronal recovery. Polyinosinic-polycytidylic acid (Poly IC) has shown neuroprotection against cerebral ischemia-reperfusion injury, but whether it regulates reactive astrogliosis and glial scar formation is not clear. METHODS: We exposed cultured astrocytes to oxygen-glucose deprivation/reoxygenation (OGD/R) and used a rat middle cerebral artery occlusion (MCAO)/reperfusion model to investigate the effects of Poly IC. Astrocyte proliferation and proliferation-related molecules were evaluated by immunostaining and Western blotting. Neurological deficit scores, infarct volumes and neuroplasticity were evaluated in rats after transient MCAO. RESULTS: In vitro, Poly IC inhibited astrocyte proliferation, upregulated Toll-like receptor 3 (TLR3) expression, upregulated interferon-ß, and downregulated interleukin-6 production. These changes were blocked by a neutralizing antibody against TLR3, suggesting that Poly IC function is TLR3-dependent. Moreover, in the MCAO model, Poly IC attenuated reactive astrogliosis, reduced brain infarction volume, and improved neurological function. In addition, Poly IC prevented MCAO-induced reductions in soma size, dendrite length, and number of dendritic bifurcations in cortical neurons of the infarct penumbra. CONCLUSIONS: By ameliorating astrogliosis-related damage, Poly IC is a potential therapeutic agent for attenuating neuronal damage and promoting recovery after brain ischemia.


Assuntos
Gliose/tratamento farmacológico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Indutores de Interferon/uso terapêutico , Poli I-C/uso terapêutico , Recuperação de Função Fisiológica/efeitos dos fármacos , Receptor 3 Toll-Like/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/ultraestrutura , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Modelos Animais de Doenças , Glucose/deficiência , Hipóxia/tratamento farmacológico , Indutores de Interferon/farmacologia , Masculino , Poli I-C/farmacologia , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão , Regulação para Cima/efeitos dos fármacos
4.
Artigo em Inglês | MEDLINE | ID: mdl-25301101

RESUMO

The hyperpolarization-activated cyclic-nucleotide-gated non-selective cation (HCN) channels play a vital role in the neurological basis underlying nervous system diseases. However, the role of HCN channels in drug addiction is not fully understood. In the present study, we investigated the expression of HCN1 and HCN2 subunits in hippocampal CA1 and the potential molecular mechanisms underlying the modulation of HCN channels in rats with chronic morphine exposure with approaches of electrophysiology, water maze, and Western blotting. We found that chronic morphine exposure (5 mg/kg, sc, for 7 days) caused an inhibition of long-term potentiation (LTP) and impairment of spatial learning and memory, which is associated with a decrease in HCN1, and an increase in HCN2 on cell membrane of hippocampal CA1 area. Additional experiments showed that the imbalance of cell membrane HCN1 and HCN2 expression under chronic morphine exposure was related to an increase in expression of TPR containing Rab8b interacting protein (TRIP8b) (1a-4) and TRIP8b (1b-2), and phosphorylation of protein kinase A (PKA) and adaptor protein 2 µ2 (AP2 µ2). Our results demonstrate the novel information that drug addiction-induced impairment of learning and memory is involved in the imbalance of HCN1 and HCN2 subunits, which is mediated by activation of TRIP8b (1a-4), TRIP8b (1b-2), PKA and AP2 µ2.


Assuntos
Região CA1 Hipocampal/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Transtornos da Memória/induzido quimicamente , Morfina/toxicidade , Entorpecentes/toxicidade , Aprendizagem Espacial/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Transtornos da Memória/patologia , Atividade Motora/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley
5.
PLoS One ; 9(6): e99526, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24914679

RESUMO

BACKGROUND: Cerebral ischemic preconditioning (IPC) protects brain against ischemic injury. Activation of Toll-like receptor 3 (TLR3) signaling can induce neuroprotective mediators, but whether astrocytic TLR3 signaling is involved in IPC-induced ischemic tolerance is not known. METHODS: IPC was modeled in mice with three brief episodes of bilateral carotid occlusion. In vitro, IPC was modeled in astrocytes by 1-h oxygen-glucose deprivation (OGD). Injury and components of the TLR3 signaling pathway were measured after a subsequent protracted ischemic event. A neutralizing antibody against TLR3 was used to evaluate the role of TLR3 signaling in ischemic tolerance. RESULTS: IPC in vivo reduced brain damage from permanent middle cerebral artery occlusion in mice and increased expression of TLR3 in cortical astrocytes. IPC also reduced damage in isolated astrocytes after 12-h OGD. In astrocytes, IPC or 12-h OGD alone increased TLR3 expression, and 12-h OGD alone increased expression of phosphorylated NFκB (pNFκB). However, IPC or 12-h OGD alone did not alter the expression of Toll/interleukin receptor domain-containing adaptor-inducing IFNß (TRIF) or phosphorylated interferon regulatory factor 3 (pIRF3). Exposure to IPC before OGD increased TRIF and pIRF3 expression but decreased pNFκB expression. Analysis of cytokines showed that 12-h OGD alone increased IFNß and IL-6 secretion; 12-h OGD preceded by IPC further increased IFNß secretion but decreased IL-6 secretion. Preconditioning with TLR3 ligand Poly I:C increased pIRF3 expression and protected astrocytes against ischemic injury; however, cells treated with a neutralizing antibody against TLR3 lacked the IPC- and Poly I:C-induced ischemic protection and augmentation of IFNß. CONCLUSIONS: The results suggest that IPC-induced ischemic tolerance is mediated by astrocytic TLR3 signaling. This reprogramming of TLR3 signaling by IPC in astrocytes may play an important role in suppression of the post-ischemic inflammatory response and thereby protect against ischemic damage. The mechanism may be via activation of the TLR3/TRIF/IRF3 signaling pathway.


Assuntos
Astrócitos/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevenção & controle , Precondicionamento Isquêmico , Receptor 3 Toll-Like/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Astrócitos/patologia , Encéfalo/patologia , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Proteína Glial Fibrilar Ácida/metabolismo , Glucose/deficiência , Infarto da Artéria Cerebral Média/patologia , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/metabolismo , Interleucina-6/metabolismo , Masculino , Camundongos , Oxigênio , Fosforilação , Poli I-C/farmacologia , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo
6.
Mol Neurobiol ; 50(2): 704-20, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24838625

RESUMO

Hyperpolarization-activated cyclic-nucleotide-gated cation nonselective (HCN) channels are involved in the pathology of nervous system diseases. HCN channels and γ-aminobutyric acid (GABA) receptors can mutually co-regulate the function of neurons in many brain areas. However, little is known about the co-regulation of HCN channels and GABA receptors in the chronic ischemic rats with possible features of vascular dementia. Protein kinase A (PKA) and TPR containing Rab8b interacting protein (TRIP8b) can modulate GABAB receptors cell surface stability and HCN channel trafficking, respectively, and adaptor-associated kinase 1 (AAK1) inhibits the function of the major TRIP8b-interacting protein adaptor protein 2 (AP2) via phosphorylating the AP2 µ2 subunit. Until now, the role of these regulatory factors in chronic cerebral hypoperfusion is unclear. In the present study, we evaluated whether and how HCN channels and GABAB receptors were pathologically altered and investigated neuroprotective effects of GABAB receptors activation and cross-talk networks between GABAB receptors and HCN channels in the hippocampal CA1 area in chronic cerebral hypoperfusion rat model. We found that cerebral hypoperfusion for 5 weeks by permanent occlusion of bilateral common carotid arteries (two-vessel occlusion, 2VO) induced marked spatial and nonspatial learning and memory deficits, significant neuronal loss and decrease in dendritic spine density, impairment of long-term potentiation (LTP) at the Schaffer collateral-CA1 synapses, and reduction of surface expression of GABAB R1, GABAB R2, and HCN1, but increase in HCN2 surface expression. Meanwhile, the protein expression of TRIP8b (1a-4), TRIP8b (1b-2), and AAK1 was significantly decreased. Baclofen, a GABAB receptor agonist, markedly improved the memory impairment and alleviated neuronal damage. Besides, baclofen attenuated the decrease of surface expression of GABAB R1, GABAB R2, and HCN1, but downregulated HCN2 surface expression. Furthermore, baclofen could restore expression of AAK1 protein and significantly increase p-PKA, TRIP8b (1a-4), TRIP8b (1b-2), and p-AP2 µ2 expression. Those findings suggested that, under chronic cerebral hypoperfusion, activation of PKA could attenuate baclofen-induced decrease in surface expression of GABAB R1 and GABAB R2, and activation of GABAB receptors not only increased the expression of TRIP8b (1a-4) and TRIP8b (1b-2) but also regulated the function of TRIP8b via AAK1 and p-AP2 µ2, which restored the balance of HCN1/HCN2 surface expression in rat hippocampal CA1 area, and thus ameliorated cognitive impairment.


Assuntos
Região CA1 Hipocampal/metabolismo , Transtornos Cognitivos/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Canais de Potássio/metabolismo , Receptores de GABA-B/metabolismo , Animais , Potenciação de Longa Duração , Masculino , Neurônios/metabolismo , Transporte Proteico/fisiologia , Ratos Sprague-Dawley , Ácido gama-Aminobutírico/metabolismo
7.
Cell Mol Neurobiol ; 33(8): 1075-86, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23975095

RESUMO

Activation of alpha2-adrenoceptors inhibits long-term potentiation and long-term depression in many brain regions. However, effectiveness and mechanism of alpha2-adrenoceptors for synaptic plasticity at the Schaffer collateral-CA1 synapses in rat in vivo is unclear. In the present study, we investigated the effects of alpha2-adrenoceptors agonist clonidine on high-frequency stimulation (HFS)-induced long-term potentiation (LTP) and paired-pulse facilitation (PPF) at the Schaffer collateral-CA1 synapse of rat hippocampus in vivo. Clonidine (0.05, 0.1 mg/kg, ip) inhibited synaptic plasticity in a dose-dependent manner, accompanying with the decreasing of aortic pressure and heart rate (HR) in anesthetized rats. Clonidine (1.25, 2.5 µg/kg, icv, 10 min before HFS) also dose-dependently inhibited synaptic plasticity, which had no remarkable effect on HR and aortic pressure. But, 20 min after HFS, administration of clonidine (2.5 µg/kg) had no effect on LTP. The inhibitory effect of clonidine (2.5 µg/kg) on LTP was completely reversed by yohimbine (18 µg/kg, icv) and ZD7288 (5 µg/kg, icv). Moreover, the inhibition was accompanied by a significant increase of the normalized PPF ratio. Furthermore, clonidine at 1 and 10 µM significantly decreased glutamate (Glu) content in the culture supernatants of hippocampal neurons, and yohimbine at 1 and 10 µM had no effect on Glu release, while it could reverse the inhibition of clonidine (1 and 10 µM) on Glu release. In conclusion, clonidine can suppress the induction of LTP at the Schaffer collateral-CA1 synapse, and the possible mechanism is that activation of presynaptic alpha2-adrenoceptors reduces the Glu release by inhibiting HCN channels.


Assuntos
Envelhecimento/fisiologia , Anestesia , Região CA1 Hipocampal/fisiologia , Clonidina/farmacologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Sinapses/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Região CA1 Hipocampal/efeitos dos fármacos , Células Cultivadas , Clonidina/administração & dosagem , Glutamatos/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Injeções Intraperitoneais , Injeções Intraventriculares , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Sinapses/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Ioimbina/administração & dosagem , Ioimbina/farmacologia
8.
Acta Pharmacol Sin ; 33(10): 1246-53, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22983393

RESUMO

AIM: To examine the neuroprotective effects of the Toll-like receptor 3 (TLR3) agonist Poly I:C in acute ischemic models in vitro and in vivo. METHODS: Primary astrocyte cultures subjected to oxygen-glucose deprivation (OGD) were used as an in vitro simulated ischemic model. Poly I:C was administrated 2 h before OGD. Cell toxicity was measured using MTT assay and LDH leakage assay. The levels of TNFα, IL-6 and interferon-ß (IFNß) in the media were measured using ELISA. Toll/interleukin receptor domain-containing adaptor-inducing IFNß (TRIF) protein levels were detected using Western blot analysis. A mouse middle cerebral artery occlusion (MCAO) model was u sed for in vivo study. The animals were administered Poly I:C (0.3 mg/kg, im) 2 h before MCAO, and examined with neurological deficit scoring and TTC staining. The levels of TNFα and IL-6 in ischemic brain were measured using ELISA. RESULTS: Pretreatment with Poly I:C (10 and 20 µg/mL) markedly attenuated OGD-induced astrocyte injury, and significantly raised the cell viability and reduced the LDH leakage. Poly I:C significantly upregulated TRIF expression accompanied by increased downstream IFNß production. Moreover, Poly I:C significantly suppressed the pro-inflammatory cytokines TNFα and IL-6 production. In mice subjected to MCAO, administration of Poly I:C significantly attenuated the neurological deficits, reduced infarction volume, and suppressed the increased levels of TNFα and IL-6 in the ischemic striatum and cortex. CONCLUSION: Poly I:C pretreatment exerts neuroprotective and anti-inflammatory effects in the simulated cerebral ischemia models, and the neuroprotection is at least in part due to the activation of the TLR3-TRIF pathway.


Assuntos
Astrócitos/efeitos dos fármacos , Ataque Isquêmico Transitório/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Poli I-C/uso terapêutico , Receptor 3 Toll-Like/agonistas , Animais , Astrócitos/metabolismo , Western Blotting , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Glucose/metabolismo , Ataque Isquêmico Transitório/metabolismo , Camundongos , Fármacos Neuroprotetores/farmacologia , Oxigênio/metabolismo , Poli I-C/farmacologia , Ratos , Ratos Sprague-Dawley
9.
J Membr Biol ; 239(3): 137-56, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21152909

RESUMO

Cardiac slow delayed rectifier (I(Ks)) channel complex consists of KCNQ1 channel and KCNE1 auxiliary subunits. The extracellular juxtamembranous region of KCNE1 is an unstructured loop that contacts multiple KCNQ1 positions in a gating-state-dependent manner. Congenital arrhythmia-related mutations have been identified in the extracellular S1-S2 linker of KCNQ1. These mutations manifest abnormal phenotypes only when coexpressed with KCNE1, pointing to the importance of proper KCNQ1/KCNE1 interactions here in I(Ks) channel function. We investigate the interactions between the KCNE1 loop (positions 36-47) and KCNQ1 S1-S2 linker (positions 140-148) by means of disulfide trapping and voltage clamp techniques. During transitions among the resting-state conformations, KCNE1 positions 36-43 make contacts with KCNQ1 positions 144, 145, and 147 in a parallel fashion. During conformational changes in the activated state, KCNE1 position 40 can make contacts with all three KCNQ1 positions, while the neighboring KCNE1 positions (36, 38, 39, and 41) can make contact with KCNQ1 position 147. Furthermore, KCNQ1 positions 143 and 146 are high-impact positions that cannot tolerate cysteine substitution. To maintain the proper I(Ks) channel function, position 143 requires a small side chain with a hydroxyl group, and position 146 requires a negatively charged side chain. These data and the proposed molecular motions provide insights into the mechanisms by which mutations in the extracellular juxtamembranous region of the I(Ks) channel impair its function.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Animais , Western Blotting , Células COS , Chlorocebus aethiops , Canal de Potássio KCNQ1/química , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Mutagênese Sítio-Dirigida , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Estrutura Secundária de Proteína , Relação Estrutura-Atividade , Xenopus laevis
10.
Cell Mol Neurobiol ; 29(3): 355-64, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19015976

RESUMO

Guattegaumerine is a natural product with characteristics of being lipophilic and reaching high concentration in the brain, but its function in the central nervous system has not yet been observed. This study was designed to evaluate the neuroprotective effects of guattegaumerine on rat primary cultured cortical neurons. Following a 24-h exposure of the cells to combined serum-starvation and hydrogen peroxide, a significant augment in neuron damage as determined by 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyl-tetrazolium bromide (MTT) assay and lactate dehydrogenase (LDH) release were observed. Preincubation of guattegaumerine dramatically improved the cell viability and inhibited LDH release. Preincubation of guattegaumerine also dramatically inhibited malondialhehyde (MDA) production and elevated the decreased total antioxidative capacity in cells caused by the combined injury. Results of flow cytometry and immunohistochemistry showed that pre-addition of guattegaumerine interrupted the apoptosis of the neurons, reversed the up regulation of the pro-apoptotic gene (Bax) and the down regulation of the anti-apoptotic gene (Bcl-2). Furthermore, guattegaumerine suppressed the increase of intracellular calcium ([Ca(2+)](i)) stimulated by either H(2)O(2) or KCl in Ca(2+)-containing extracellular solutions, and high concentration of 2.5 microM guattegaumerine also suppressed the increase of [Ca(2+)](i) induced by H(2)O(2) in Ca(2+)-free solution. These observations suggested that guattegaumerine may possess potential protection against oxidative stress injury, which might be beneficial for neurons.


Assuntos
Córtex Cerebral/citologia , Citoproteção/efeitos dos fármacos , Peróxido de Hidrogênio/toxicidade , Isoquinolinas/farmacologia , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Soro , Animais , Animais Recém-Nascidos , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Isoquinolinas/química , L-Lactato Desidrogenase/metabolismo , Malondialdeído/metabolismo , Neurônios/citologia , Cloreto de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Proteína X Associada a bcl-2/metabolismo
11.
Yao Xue Xue Bao ; 41(6): 565-71, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16927834

RESUMO

AIM: To study the effect of ZD7288 on synaptic transmission in the pathway from perforant pathway (PP) fibers to CA3 region in rat hippocampus. METHODS: The extracellular recording technique in vivo was used to record the CA3 region field potentials. High-performance liquid chromatography (HPLC) with fluorescence detection was applied to measure the content of amino acids in hippocampal tissues. The effect of ZD7288 and CsCl on the amplitudes of population spike (PS) in CA3 region evoked by stimulation (0.5 Hz) of the perforant pathway (PP) fibers, and the content of amino acids in hippocampal tissue were observed. RESULTS: Microinjection of ZD7288 (20, 100 and 200 nmol) and CsCl (1, 5 and 10 micromol) into CA3 region decreased the population spike (PS) amplitudes in a dose-dependent manner. The inhibitory effects appeared at 5 min after microinjection and lasted at least 90 min. In those rats treated with ZD7288 (100 nmol), the contents of glutamate, aspartate, glycine and GABA decreased significantly as compared to those of saline control (all P < 0.01, except P < 0.05 for that of glycine). A similar decrease in the contents of amino acids was observed when the rats were microinjected with CsCl (5 micromol). CONCLUSION; ZD7288 could obviously inhibit synaptic transmission in the pathway from PP fibers to CA3 region in rat hippocampus, and this action of ZD7288 may be associated with altered contents of amino acids.


Assuntos
Aminoácidos/metabolismo , Hipocampo/metabolismo , Pirimidinas/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Animais , Césio/farmacologia , Cloretos/farmacologia , Relação Dose-Resposta a Droga , Potenciais Evocados , Hipocampo/fisiologia , Masculino , Microinjeções , Via Perfurante/fisiologia , Pirimidinas/administração & dosagem , Ratos , Ratos Sprague-Dawley
12.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 26 Suppl: 7-10, 2006 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-17569334

RESUMO

OBJECTIVE: To observe the protective effect of Kang Naoxueshuan Tablet (KNT) on ischemic brain injury in rats, and explore its possible mechanism. METHODS: Rats were administrated with KNT twice per day for successive 14 days. Rat model of acute focal cerebral ischemia was established by middle cerebral artery occlusion (MCAO) with a nylon suture inserted through the right internal carotid artery to occlude the beginning of middle cerebral artery. After 24 hrs MCAO, the neurological deficit and the volume of cerebral infarct were observed, and the effect of KNT on the thrombosis of rats in vitro, platelet aggregation and blood viscosity were also determined. RESULTS: KNT could alleviate volume of cerebral infarct caused by focal cerebral ischemia in a dose-dependent manner and improve neurological symptoms. The volume of cerebral infarct was 11. 18 +/- 3. 35% , 14. 60 +/- 7.00% and 15. 37 +/- 7. 21% in the high, middle and low-dose groups, respectively, and they were decreased 59. 36% , 46. 93% and 44. 13% than that in the model group 27. 51 +/- 4. 71% (P <0. 01 ). The wet and dry weigh of thrombosis in vitro of the three different dose groups were significantly decreased, and they were significantly different than that of the model group (P <0. 05, P <0. 01). KNT could significantly inhibit platelet aggregation induced by ADP and decrease blood viscosity, but it had no effect on plasma viscosity and hematocrit. CONCLUSION: KNT has significant protective effect on ischemia, the mechanism is relateed to the improvement of blood viscosity and inhibiti on of platelet aggregation. But the exact mechanisms need to be probed into deeply.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Medicina Tradicional Chinesa , Inibidores da Agregação Plaquetária/uso terapêutico , Substâncias Protetoras/uso terapêutico , Animais , Viscosidade Sanguínea/efeitos dos fármacos , Isquemia Encefálica/sangue , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/administração & dosagem , Substâncias Protetoras/administração & dosagem , Ratos , Comprimidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...