Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Med ; 42(5): 2750-2762, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30226562

RESUMO

GPR17 is a G (i)-coupled dual receptor, linked to P2Y and CysLT receptors stimulated by uracil nucleotides and cysteinyl leukotrienes, respectively. Recent evidence has demonstrated that GPR17 inhibition ameliorates the progression of cerebral ischemic injury by regulating neuronal death and microglial activation. The present study aimed to assess the detailed regulatory roles of this receptor in oxygen­glucose deprivation/recovery (OGD/R)­induced ischemia­like injury in vitro and explore the underlying mechanism. The results demonstrated that OGD/R induced ischemic neuronal injury and microglial activation, including enhanced phagocytosis and increased inflammatory cytokine release in neuron­glial mixed cultures of cortical cells. GPR17 upregulation during OGD/R was spatially and temporally correlated with neuronal injury and microglial activation. In addition, GPR17 knockdown inhibited OGD/R­induced responses in neuron­glial mixed cultures. GPR17 knockdown also attenuated cell injury induced by the agonist leukotriene D4 (LTD4) or uridine 5'­diphosphate (UDP) in neuron­glial mixed cultures. However, GPR17 knockdown did not affect OGD/R­induced ischemic neuronal injury in primary cultures of neurons. In primary astrocyte cultures, neither GPR17 nor OGD/R induced injury. By contrast, GPR17 knockdown ameliorated OGD/R­induced microglial activation, boosting phagocytosis and inflammatory cytokine release in primary microglia cultures. Finally, the results demonstrated that the conditioned medium of microglia pretreated with OGD/R induced neuronal death, and the neuronal injury was significantly inhibited by GPR17 knockdown. These findings suggested that GPR17 may mediate ischemia­like neuronal injury and microglial activation in vitro; however, the protective effects on ischemic neuronal injury might depend upon microglial activation. Whether GPR17 regulates neuronal injury mediated by oligodendrocyte linkage remains to be investigated.


Assuntos
Citocinas/imunologia , Microglia/patologia , Neurônios/patologia , Receptores Acoplados a Proteínas G/imunologia , Traumatismo por Reperfusão/patologia , Animais , Astrócitos/imunologia , Astrócitos/metabolismo , Astrócitos/patologia , Isquemia Encefálica/genética , Isquemia Encefálica/imunologia , Isquemia Encefálica/patologia , Morte Celular , Células Cultivadas , Microglia/imunologia , Microglia/metabolismo , Neurônios/imunologia , Neurônios/metabolismo , Fagocitose , Interferência de RNA , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/genética , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/imunologia , Regulação para Cima
2.
Neuroscience ; 356: 193-206, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28528966

RESUMO

Nicotinamide phosphoribosyltransferase (NAMPT) is an important neuroprotective factor in cerebral ischemia, and it has been reported that NAMPT inhibitors can aggravate neuronal injury in the acute phase. However, because it is a cytokine, NAMPT participates in many inflammatory diseases in the peripheral system, and its inhibitors have therapeutic effects. Following cerebral ischemia, the peripheral and resident inflammatory and immune cells produce many pro-inflammatory mediators in the ischemic area, which induce neuroinflammation and impair the brain. However, the effects of NAMPT inhibitors in the neuroinflammation after ischemic brain injury remain unknown. Here, we found that FK866, a potent NAMPT inhibitor, decreased the level of TNF-α, NAMPT and IL-6 in the ischemic brain tissue one day after middle-cerebral-artery occlusion and reperfusion (MCAO/R), improved neurological dysfunction, decreased infarct volume and neuronal loss, and inhibited microgliosis and astrogliosis 14days after MCAO/R. The expression of NAMPT protein was induced in Iba1-positive microglia/macrophages in the ischemia core 14days after MCAO/R. In vitro studies show that oxygen-glucose deprivation and recovery (OGD/R) activate microglia. Activated microglia increased the activity of NF-κB, increased the mRNA synthesis of TNF-α, NAMPT and IL-6, and increased the secretion of TNF-α, NAMPT and IL-6. On the other hand, NAMPT can act synergistically with other cytokines and activate microglia. FK866 strongly inhibited these changes and alleviated OGD/R-induced activation of microglia. As such, NAMPT is a crucial determinant of cellular inflammation after cerebral ischemia. NAMPT inhibitors are novel compounds to protect neuronal injury from ischemia via anti-inflammatory effects.


Assuntos
Encéfalo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Nicotinamida Fosforribosiltransferase/efeitos dos fármacos , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Isquemia/tratamento farmacológico , Isquemia/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Microglia/efeitos dos fármacos , NF-kappa B/metabolismo , Neurônios/metabolismo , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo
3.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 43(3): 257-64, 2014 05.
Artigo em Chinês | MEDLINE | ID: mdl-24998647

RESUMO

OBJECTIVE: To investigate the antioxidative effects of two cysteinyl leukotriene receptors antagonists (CysLT1R and CysLT2R) montelukast and HAMI 3379 on ischemic injury of rat cortical neurons in vitro. METHODS: Cultured rat cortical neurons were pretreated with CysLT1R antagonist montelukast and CysLT2R antagonist HAMI 3379, and then exposed to oxygen-glucose deprivation/recovery (OGD/R)or H2O2. Reactive oxygen species (ROS) mitochondrial membrane potential (MMP) depolarization, neuronal viability and lactate dehydrogenase (LDH) release were determined. Meanwhile, RNA interference was used to inhibit the expression of CysLT1R and CysLT2R,and the effects were observed. RESULTS: ROS production in neurons was significantly increased after 1 h OGD, which reached the peak at 30 min and lasted for 1.5 h after recovery. Montelukast and HAMI 3379 at 0.01-1µmol/L moderately decreased OGD/R-induced ROS production (P<0.05). Montelukast mildly attenuated OGD/R-induced MMP depolarization (P<0.05),but HAMI 3379 had no effect. H2O2 reduced neuronal viability and increased LDH release, namely inducing neuronal injury. Montelukast and HAMI 3379 at 0.1-1µmol/L moderately attenuated H2O2-induced neuronal injury (P<0.05). However, both CysLT1R siRNA and CysLT2R shRNA did not significantly affect the responses mentioned above. CONCLUSION: In ischemic neuronal injury, montelukast and HAMI 3379 exert a moderate antioxidative effect, and this effect may be receptor-independent.


Assuntos
Acetatos/farmacologia , Antioxidantes/farmacologia , Ácidos Cicloexanocarboxílicos/farmacologia , Neurônios/efeitos dos fármacos , Ácidos Ftálicos/farmacologia , Quinolinas/farmacologia , Animais , Hipóxia Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Ciclopropanos , Antagonistas de Leucotrienos/farmacologia , Neurônios/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Sulfetos
4.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 43(3): 287-92, 2014 05.
Artigo em Chinês | MEDLINE | ID: mdl-24998651

RESUMO

OBJECTIVE: To investigate the effects of cysteinyl leukotriene (CysLT) receptor agonist leukotriene D4 (LTD4) on proliferation and migration in lung epithelial A549 cells. METHODS: The expression of CysLT1 receptor and CysLT2 receptor was determined by immunofluoresence staining in A549 cells. A549 cells were treated with LTD4 (0.01-100 nmol/L) for 24-72 h. Cell viability was detected by MTT reduction assay. Cell migration was determined by modified scratch and healing model. RESULTS: In A549 cells, CysLT1 receptor and CysLT2 receptor were mainly expressed in the cytoplasm, membrane and few in the nuclei. The treatment of LTD4 (0.01-100 nmol/L) for 24-72 h caused no effect on cell viability (Ps>0.05); when A549 cells were treated with 100 nmol/L LTD4 for 24, 48 and 72 h the cell viability was (103.00±4.46)%,(107.00±9.45)% and (105.00±9.02)% of control, respectively (Ps>0.05). The migration rate of A549 cells after scratching during the first 24 h was markedly greater than that during the second and third 24 h in the same concentration groups; however, no significant difference in migration rate was noticed when the cells were treated with different concentrations of LTD4 (0.01-100 nmol/L)(Ps>0.05). The migration of A549 cells was 1.15-fold, 1.21-fold and 1.06-fold of that of control when the cells were treated with 100 nmol/L LTD4 for 24, 48 and 72 h, respectively (Ps>0.05). CONCLUSION: The proliferation and migration of A549 cells are not changed when treated with 0.01-100 nmol LTD4 for up to 72h.


Assuntos
Células Epiteliais/citologia , Leucotrieno D4/farmacologia , Alvéolos Pulmonares/citologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Humanos
5.
Acta Pharmacol Sin ; 35(1): 33-40, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24141567

RESUMO

AIM: To investigate the roles of cysteinyl leukotriene receptors CysLT1R and CysLT2R in leukotriene D4 (LTD4)-induced activation of microglial cells in vitro. METHODS: Mouse microglial cell line BV2 was transfected with pcDNA3.1(+)-hCysLT1R or pcDNA3.1(+)-hCysLT2R. The expression of relevant mRNAs and proteins in the cells was detected using RT-PCR and Western blotting, respectively. Phagocytosis was determined with flow cytometry analysis. The release of interleukin-1ß (IL-1ß) from the cells was measured using an ELISA assay. RESULTS: The expression of CysLT1R or CysLT2R was considerably increased in the transfected BV2 cells, and the receptors were mainly distributed in the plasma membrane and cytosol. Treatment of the cells expressing CysLT1R or CysLT2R with CysLT receptor agonist LTD4 (0.1-100 nmol/L) concentration-dependently enhanced the phagocytosis, and increased mRNA expression and release of IL-1ß. Moreover, the responses of hCysLT1R-BV2 cells to LTD4 were significantly larger than those of hCysLT2R-BV2 or WT-BV2 cells. Pretreatment of hCysLT1R-BV2 cells with the selective CysLT1R antagonist montelukast (1 µmol/L) significantly blocked LTD4-induced phagocytosis as well as the mRNA expression and release of IL-1ß, whereas the selective CysLT2R antagonist HAMI 3379 (1 µmol/L) had no such effects. CONCLUSION: CysLT1R mediates LTD4-induced activation of BV2 cells, suggesting that CysLT1R antagonists may exert anti-inflammatory activity in brain diseases.


Assuntos
Leucotrieno D4/farmacologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Receptores de Leucotrienos/agonistas , Receptores de Leucotrienos/fisiologia , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Humanos , Camundongos
6.
J Pharmacol Exp Ther ; 346(2): 328-41, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23750020

RESUMO

The cysteinyl leukotrienes (CysLTs) are inflammatory mediators closely associated with neuronal injury after brain ischemia through the activation of their receptors, CysLT1R and CysLT2R. Here we investigated the involvement of both receptors in oxygen-glucose deprivation/recovery (OGD/R)-induced ischemic neuronal injury and the effect of the novel CysLT2R antagonist HAMI 3379 [3-({[(1S,3S)-3- carboxycyclohexyl]amino}carbonyl)-4-(3-{4-[4-(cyclo-hexyloxy)butoxy]phenyl}propoxy)benzoic acid] in comparison with the CysLT1R antagonist montelukast. In primary neurons, neither the nonselective agonist leukotriene D4 (LTD4) nor the CysLT2R agonist N-methyl-leukotriene C4 (NMLTC4) induced neuronal injury, and HAMI 3379 did not affect OGD/R-induced neuronal injury. However, in addition to OGD/R, LTD4 and NMLTC4 induced cell injury and neuronal loss in mixed cultures of cortical cells, and neuronal loss and necrosis in neuron-microglial cocultures. Moreover, they induced phagocytosis and cytokine release (interleukin-1ß and tumor necrosis factor-α) from primary microglia, and conditioned medium from the treated microglia induced neuronal necrosis. HAMI 3379 inhibited all of these responses, and its effects were the same as those of CysLT2R interference by CysLT2R short hairpin RNA, indicating CysLT2R dependence. In comparison, montelukast moderately inhibited OGD/R-induced primary neuronal injury and most OGD/R- and LTD4-induced (but not NMLTC4-induced) responses in mixed cultures, cocultures, and microglia. The effects of montelukast were both dependent and independent of CysLT1Rs because interference by CysLT1R small interfering RNA had limited effects on neuronal injury in neuron-microglial cocultures and on cytokine release from microglia. Our findings indicated that HAMI 3379 effectively blocked CysLT2R-mediated microglial activation, thereby indirectly attenuating ischemic neuronal injury. Therefore, CysLT2R antagonists may represent a new type of therapeutic agent in the treatment of ischemic stroke.


Assuntos
Ácidos Cicloexanocarboxílicos/farmacologia , Antagonistas de Leucotrienos/farmacologia , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ácidos Ftálicos/farmacologia , Receptores de Leucotrienos/metabolismo , Acetatos/farmacologia , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Hipóxia Celular , Células Cultivadas , Córtex Cerebral/citologia , Técnicas de Cocultura , Ciclopropanos , Citocinas/metabolismo , Feminino , Glucose/metabolismo , Masculino , Microglia/metabolismo , Microglia/patologia , Necrose , Neurônios/metabolismo , Neurônios/patologia , Oxigênio/metabolismo , Fagocitose , Cultura Primária de Células , Quinolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Leucotrienos/agonistas , Sulfetos
7.
Neurosci Bull ; 28(6): 680-92, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23132680

RESUMO

OBJECTIVE: Aquaporin-4 (AQP4), the main water channel protein in the brain, plays a critical role in water homeostasis and brain edema. Here, we investigated its role in the inflammatory responses after focal cerebral ischemia. METHODS: In AQP4-knockout (KO) and wild-type mice, focal cerebral ischemia was induced by 30 min of middle cerebral arterial occlusion (MCAO). Ischemic neuronal injury and cellular inflammatory responses, as well as the expression and localization of cysteinyl leukotriene CysLT(2) and CysLT(1) receptors, were determined at 24 and 72 h after MCAO. RESULTS: AQP4-KO mice showed more neuronal loss, more severe microglial activation and neutrophil infiltration, but less astrocyte proliferation in the brain after MCAO than wild-type mice. In addition, the protein levels of both CysLT(1) and CysLT(2) receptors were up-regulated in the ischemic brain, and the up-regulation was more pronounced in AQP4-KO mice. The CysLT(1) and CysLT(2) receptors were primarily localized in neurons, microglia and neutrophils; those localized in microglia and neutrophils were enhanced in AQP4-KO mice. CONCLUSION: AQP4 may play an inhibitory role in postischemic inflammation.


Assuntos
Aquaporina 4/deficiência , Isquemia Encefálica/metabolismo , Inflamação/metabolismo , Receptores de Leucotrienos/biossíntese , Animais , Aquaporina 4/genética , Astrócitos/metabolismo , Western Blotting , Isquemia Encefálica/patologia , Contagem de Células , Imuno-Histoquímica , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Inflamação/patologia , Leucócitos/metabolismo , Camundongos , Camundongos Knockout , Microglia/fisiologia , Neutrófilos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Regulação para Cima , Intoxicação por Água/metabolismo
8.
Acta Pharmacol Sin ; 33(12): 1511-7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23085741

RESUMO

AIM: Cysteinyl leukotriene receptor 1 (CysLT(1) receptor) is located in epithelial cells, and translocates from the plasma membrane to the nucleus in a ligand-dependent manner. Here, we investigated whether CysLT(1) receptors translocated to the nucleus in endothelial cells after ischemic insult in vitro and whether it was involved in ischemic injury to endothelial cells. METHODS: EA.hy926 cell line, derived from human umbilical vein endothelial cells, was subjected to oxygen-glucose deprivation (OGD). The expression and distribution of CysLT(1) receptors were detected by immunofluorescent staining, immunogold labeling and immunoblotting analyses. Cell viability was evaluated using MTT reduction assay. Necrosis and apoptosis were determined by double fluorescent staining with propidium iodide and Hoechst 33342. RESULTS: CysLT(1) receptors were primarily distributed in the cytoplasm and nucleus in EA.hy926 cells, and few was found in the cell membrane. OGD induced the translocation of CysLT(1) receptors from the cytoplasm to the nucleus in a time-depen dent manner, with a peak reached at 6 h. OGD-induced nuclear translocation of CysLT(1) receptors was inhibited by pretreatment with the CysLT(1) receptor antagonist pranlukast (10 µmol/L), or by preincubation with NLS-pep, a peptide corresponding to the nuclear localization sequence of CysLT(1) receptor (10 µg/mL). However, zileuton, an inhibitor of 5-lipoxygenase that was a key enzyme in cysteinyl leukotriene generation, did not inhibit the nuclear translocation of CysLT(1) receptors. Moreover, preincubation with NLS-pep (0.4 µg/mL) significantly ameliorated OGD-induced cell viability reduction and necrosis. CONCLUSION: CysLT(1) receptors in endothelial cells translocate to the nucleus in a ligand-independent manner after ischemic insult in vitro, and it is involved in the ischemic injury.


Assuntos
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Células Endoteliais/metabolismo , Glucose/metabolismo , Oxigênio/metabolismo , Receptores de Leucotrienos/metabolismo , Apoptose/efeitos dos fármacos , Isquemia Encefálica/metabolismo , Técnicas de Cultura de Células , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cromonas/farmacologia , Células Endoteliais/efeitos dos fármacos , Humanos , Hidroxiureia/análogos & derivados , Hidroxiureia/farmacologia , Ligantes , Modelos Biológicos , Sinais de Localização Nuclear/farmacologia , Transporte Proteico
9.
Brain Res ; 1484: 57-67, 2012 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-23000196

RESUMO

Cysteinyl leukotrienes (CysLTs) induce inflammatory responses by activating their receptors, CysLT(1)R and CysLT(2)R. We recently reported that CysLT(2)R is involved in neuronal injury, astrocytosis and microgliosis after focal cerebral ischemia in rats. Here, we determined whether HAMI 3379, a selective CysLT(2)R antagonist, protects against acute brain injury after focal cerebral ischemia in rats. We induced transient focal cerebral ischemia by 30 min of middle cerebral artery occlusion (MCAO), followed by 24h of reperfusion. HAMI 3379 (1, 10 or 100 ng) was injected intracerebroventricularly (i.c.v.) 30 min before MCAO, and the CysLT(1)R antagonist pranlukast (0.1mg/kg, i.p.) was used as a positive control. HAMI 3379 at 10 and 100 ng (but not at 1 ng) attenuated the neurological deficits, and reduced infarct volume, brain edema, IgG exudation, neuronal degeneration and neuronal loss. This protective effect was similar to that of pranlukast. Thus, HAMI 3339 at 10-100 ng i.c.v. is neuroprotective against acute brain injury after focal cerebral ischemia in rats. These findings suggest therapeutic potential for CysLT(2)R antagonists in the treatment of ischemic stroke.


Assuntos
Lesões Encefálicas/prevenção & controle , Ácidos Cicloexanocarboxílicos/administração & dosagem , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fármacos Neuroprotetores/administração & dosagem , Ácidos Ftálicos/administração & dosagem , Receptores de Leucotrienos/metabolismo , Animais , Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/patologia , Injeções Intraventriculares , Antagonistas de Leucotrienos/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Leucotrienos/efeitos dos fármacos
10.
J Neuroinflammation ; 9: 145, 2012 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-22734808

RESUMO

BACKGROUND: Transforming growth factor-ß 1 (TGF-ß 1) is an important regulator of cell migration and plays a role in the scarring response in injured brain. It is also reported that 5-lipoxygenase (5-LOX) and its products, cysteinyl leukotrienes (CysLTs, namely LTC4, LTD4 and LTE4), as well as cysteinyl leukotriene receptor 1 (CysLT1R) are closely associated with astrocyte proliferation and glial scar formation after brain injury. However, how these molecules act on astrocyte migration, an initial step of the scarring response, is unknown. To clarify this, we determined the roles of 5-LOX and CysLT1R in TGF-ß 1-induced astrocyte migration. METHODS: In primary cultures of rat astrocytes, the effects of TGF-ß 1 and CysLT receptor agonists on migration and proliferation were assayed, and the expression of 5-LOX, CysLT receptors and TGF-ß1 was detected. 5-LOX activation was analyzed by measuring its products (CysLTs) and applying its inhibitor. The role of CysLT1R was investigated by applying CysLT receptor antagonists and CysLT1R knockdown by small interfering RNA (siRNA). TGF-ß 1 release was assayed as well. RESULTS: TGF-ß 1-induced astrocyte migration was potentiated by LTD4, but attenuated by the 5-LOX inhibitor zileuton and the CysLT1R antagonist montelukast. The non-selective agonist LTD4 at 0.1 to 10 nM also induced a mild migration; however, the selective agonist N-methyl-LTC4 and the selective antagonist Bay cysLT2 for CysLT2R had no effects. Moreover, CysLT1R siRNA inhibited TGF-ß 1- and LTD4-induced astrocyte migration by down-regulating the expression of this receptor. However, TGF-ß 1 and LTD4 at various concentrations did not affect astrocyte proliferation 24 h after exposure. On the other hand, TGF-ß 1 increased 5-LOX expression and the production of CysLTs, and up-regulated CysLT1R (not CysLT2R), while LTD4 and N-methyl-LTC4 did not affect TGF-ß 1 expression and release. CONCLUSIONS: TGF-ß 1-induced astrocyte migration is, at least in part, mediated by enhanced endogenous CysLTs through activating CysLT1R. These findings indicate that the interaction between the cytokine TGF-ß 1 and the pro-inflammatory mediators CysLTs in the regulation of astrocyte function is relevant to glial scar formation.


Assuntos
Araquidonato 5-Lipoxigenase/metabolismo , Astrócitos/metabolismo , Movimento Celular/imunologia , Movimento Celular/fisiologia , Receptores de Leucotrienos/metabolismo , Fator de Crescimento Transformador beta1/fisiologia , Animais , Animais Recém-Nascidos , Araquidonato 5-Lipoxigenase/fisiologia , Astrócitos/citologia , Ativação Enzimática/fisiologia , Leucotrieno D4/farmacologia , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Receptores de Leucotrienos/fisiologia , Fator de Crescimento Transformador beta1/farmacologia
11.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 41(3): 259-66, 2012 05.
Artigo em Chinês | MEDLINE | ID: mdl-22723160

RESUMO

OBJECTIVE: To determine the effect of montelukast, a cysteinyl leukotriene receptor 1 antagonist, on morphological changes in rat neurons after ischemic injury. METHODS: The in vivo ischemia injury was induced by oxygen-glucose deprivation (OGD) for 2 h and reperfusion (R) for 24 h (OGD/R) in rat neurons primary culture and mixed cortex culture. In the presence or absence of various concentrations of montelukast, neuron number, area of neuron, number of neuritis per neuron, branch number of primary neuritis and primary neurite length were determined for evaluating morphological changes in neurons. RESULTS: OGD/R significantly reduced neuron number, and altered neuron morphology. In cortical neuron cultures, montelukast (0.0001-1 µmol/L) attenuated OGD/R-induced reduction in neuron number, and inhibited OGD/R-induced increase in branch number of primary neuritis. In the mixed cultures, montelukast (0.0001-0.1 µmol/L) increased the primary neurite length, and reduced number of neuritis and branch number of primary neurite after OGD/R. CONCLUSION: Montelukast has a protective effect on ischemic injury in neurons.


Assuntos
Acetatos/farmacologia , Neurônios/patologia , Quinolinas/farmacologia , Animais , Animais Recém-Nascidos , Hipóxia Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ciclopropanos , Glucose/farmacologia , Antagonistas de Leucotrienos/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley , Sulfetos
12.
Eur J Pharmacol ; 674(2-3): 163-70, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22119381

RESUMO

Nicotinamide phosphoribosyltransferase (NAMPT) is a key enzyme in the salvaging pathway for the synthesis of nicotinamide adenine dinucleotide (NAD) that is involved in cell metabolism and proliferation. NAMPT is normally absent in astrocyte but highly expressed in glioblastoma, suggesting that it may promote cell survival through synthesizing more NAD. In this report, we evaluated the effect of APO866, a potent inhibitor of NAMPT against C6 glioblastoma. We found that APO866 inhibited the growth of C6 glioblastoma cells with IC(50) in nano-molar range. APO866 depleted intracellular NAD, caused marked inhibition of ERK activation and induced G2/M cell-cycle arrest. The effects by APO866 were abrogated by nicotinamide mononucleotide (NMN), the direct product of NAMPT. Administration of U0126, an ERK1/2 inhibitor, inhibited cell growth but displayed no synergistic effect with APO866. Taken together, our results indicated that APO866 is a potent growth inhibitor against glioblastoma through targeting NAMPT.


Assuntos
Acrilamidas/farmacologia , Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Glioblastoma/patologia , Nicotinamida Fosforribosiltransferase/antagonistas & inibidores , Piperidinas/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NAD/metabolismo , Nicotinamida Fosforribosiltransferase/metabolismo , Ratos
13.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 40(2): 139-44, 2011 03.
Artigo em Chinês | MEDLINE | ID: mdl-21488208

RESUMO

OBJECTIVE: To investigate the role of cysteinyl leukotriene (CysLT) receptors in the differentiation of rat glioma C6 cells. METHODS: Rat glioma C6 cells were treated with the agonist LTD(4), the CysLT(1) receptor antagonist montelukast and the differentiation inducer forskolin. Cell morphology and GFAP protein expression were determined after treatments. RESULT: Forskolin (10 µmol/L) induced morphological changes and GFAP protein expression (cell differentiation) in C6 cells, but LTD(4) (0.1-100 nmol/L) did not induce these changes. Montelukast (1 µmol/L) alone did not affect C6 cell differentiation, while it induced the differentiation when combined with the LTD(4) (100 nmol/L). CONCLUSION: The CysLT(2) receptor may modulate the differentiation of rat glioma C6 cells.


Assuntos
Glioma/patologia , Antagonistas de Leucotrienos/farmacologia , Receptores de Leucotrienos/agonistas , Acetatos/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Colforsina/farmacologia , Ciclopropanos , Cisteína , Glioma/metabolismo , Leucotrieno D4/farmacologia , Leucotrienos , Quinolinas/farmacologia , Ratos , Sulfetos
14.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 40(2): 131-8, 2011 03.
Artigo em Chinês | MEDLINE | ID: mdl-21488207

RESUMO

OBJECTIVE: To prepare and identify a polyclonal antibody (pAb) against (mouse) cysteinyl leukotriene receptor 1 (CysLT(1)) and to investigate the changes of CysLT(1) receptor expression in BV2 microglial cells after rotenone treatment. METHODS: Rabbits were immunized with KLH-coupled CysLT(1) peptide to prepare the pAb. The titer of the pAb in rabbit plasma was detected by ELISA method, and the specificity of the pAb was tested by antigen blockade. After BV2 cells were treated with rotenone (0.01-1 µmol/L) for 24 h, the expression of CysLT(1) was determined by immunostaining, Western blotting and RT-PCR. RESULT: The pAb showed a titer of 1/32728, and was not cross-reacted with antigens of CysLT(2) receptor and GPR17. Immunostaining, Western blotting and RT-PCR analysis showed the expression of CysLT(1) receptor in BV2 microglia. Rotenone at 1µmol/L significantly induced an increased expression of CysLT(1) receptor. CONCLUSION: The prepared CysLT(1) receptor polyclonal antibody has a high titer and high specificity to meet testing requirements of Western blotting and immunostaining; CysLT(1) is associated with rotenone-induced injury of BV2 microglial cells.


Assuntos
Microglia/metabolismo , Receptores de Leucotrienos/metabolismo , Rotenona/farmacologia , Animais , Células Cultivadas , Masculino , Camundongos , Microglia/efeitos dos fármacos , Microglia/patologia , Coelhos , Receptores de Leucotrienos/imunologia
15.
J Pharm Pharmacol ; 63(4): 550-7, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21401607

RESUMO

OBJECTIVES: Previously we demonstrated the neuroprotective effect of montelukast, a cysteinyl leukotriene receptor-1 (CysLT(1) ) antagonist, on acute brain injury after focal cerebral ischaemia in mice. In this study, we have determined its effect on chronic brain injury after focal cerebral ischaemia in mice and rats. METHODS: After transient focal cerebral ischaemia was induced by middle cerebral artery occlusion, montelukast was intraperitoneally injected in mice or orally administered to rats for five days. Behavioural dysfunction, brain infarct volume, brain atrophy and neuron loss were determined to evaluate brain lesions. KEY FINDINGS: Montelukast (0.1 mg/kg) attenuated behavioural dysfunction, brain infarct volume, brain atrophy and neuron loss in mice, which was similar to pranlukast, another CysLT(1) receptor antagonist. Oral montelukast (0.5 mg/kg) was effective in rats and was more effective than edaravone, a free radical scavenger. CONCLUSION: Montelukast protected mice and rats against chronic brain injury after focal cerebral ischaemia, supporting the therapeutic potential of CysLT(1) receptor antagonists.


Assuntos
Acetatos/uso terapêutico , Antipirina/análogos & derivados , Lesão Encefálica Crônica/patologia , Lesão Encefálica Crônica/prevenção & controle , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/patologia , Degeneração Neural/tratamento farmacológico , Quinolinas/uso terapêutico , Acetatos/administração & dosagem , Acetatos/farmacologia , Administração Oral , Animais , Antipirina/uso terapêutico , Atrofia/prevenção & controle , Comportamento Animal/efeitos dos fármacos , Lesão Encefálica Crônica/complicações , Cromonas/uso terapêutico , Ciclopropanos , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Edaravone , Sequestradores de Radicais Livres/uso terapêutico , Injeções Intraperitoneais , Antagonistas de Leucotrienos/farmacologia , Antagonistas de Leucotrienos/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos ICR , Quinolinas/administração & dosagem , Quinolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Sulfetos
16.
J Cardiovasc Pharmacol ; 57(4): 479-88, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21283018

RESUMO

OBJECTIVE: Recently, we reported that pranlukast, an antagonist of cysteinyl leukotriene receptor 1, attenuates ischemic injury in endothelial cells by decreasing reactive oxygen species (ROS) production and inhibiting nuclear factor-κB activation in a leukotriene-independent manner. In this study, we investigated the effect of pranlukast on oxidative stress injury induced by hydrogen peroxide (H2O2) in EA.hy926 cells, a human endothelial cell line, and the possible mechanisms. METHODS AND RESULTS: We found that H2O2 reduced cell viability and increased lactate dehydrogenase release in a concentration- and time-dependent manner. Necrosis was the main death mode, and the necrotic rate increased 32% after exposure to 220 µM H2O2 for 4 hours. Pretreatment with pranlukast significantly ameliorated the reduced viability and the increased lactate dehydrogenase release and necrosis after exposure to H2O2. We next examined the mechanisms underlying the antinecrotic effects of pranlukast. The results showed that pranlukast attenuated excessive ROS production and ameliorated the reduced superoxide dismuase and glutathione peroxidase activity in EA.hy926 cells exposed to H2O2. Pranlukast also inhibited the collapse of mitochondrial membrane potential (MMP) induced by H2O2. Inhibition of ROS production by N-acetyl-l-cysteine, a powerful antioxidant, reduced MMP collapse and necrosis. Inhibition of MMP collapse by cyclosporine A, a mitochondrial permeability transition inhibitor, attenuated necrosis but failed to reduce ROS production. In addition, we found no expression of 5-lipoxygenase in EA.hy926 cells and zileuton, a 5-lipoxygenase inhibitor, did not affect the cellular injury induced by H2O2. CONCLUSION: Pranlukast protects endothelial cells from H2O2-induced necrosis by inhibiting ROS-mediated collapse of mitochondrial membrane potential, and this is leukotriene-independent.


Assuntos
Cromonas/farmacologia , Antagonistas de Leucotrienos/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Peróxido de Hidrogênio/administração & dosagem , Peróxido de Hidrogênio/toxicidade , L-Lactato Desidrogenase/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Necrose/tratamento farmacológico , Necrose/patologia , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
17.
Life Sci ; 88(1-2): 50-6, 2011 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-21055410

RESUMO

AIMS: We previously reported that cysteinyl leukotriene receptor 2 (CysLT(2)) mediates ischemic astrocyte injury, and leukotriene D(4)-activated CysLT(2) receptor up-regulates the water channel aquaporin 4 (AQP4). Here we investigated the mechanism underlying CysLT(2) receptor-mediated ischemic astrocyte injury induced by 4-h oxygen-glucose deprivation and 24-h recovery (OGD/R). MAIN METHODS: Primary cultures of rat astrocytes were treated by OGD/R to construct the cell injury model. AQP4 expression was inhibited by small interfering RNA (siRNA). The expressions of AQP4 and CysLTs receptors, and the MAPK signaling pathway were determined. KEY FINDINGS: OGD/R induced astrocyte injury, and increased expression of the CysLT(2) (but not CysLT(1)) receptor and AQP4. OGD/R-induced cell injury and AQP4 up-regulation were inhibited by a CysLT(2) receptor antagonist (Bay cysLT2) and a non-selective CysLT receptor antagonist (Bay u9773), but not by a CysLT(1) receptor antagonist (montelukast). Knockdown of AQP4 by siRNA attenuated OGD/R injury. Furthermore, OGD/R increased phosphorylation of ERK1/2 and p38, whose inhibitors relieved the cell injury and AQP4 up-regulation. SIGNIFICANCE: The CysLT(2) receptor mediates AQP4 up-regulation in astrocytes, and up-regulated AQP4 leads to OGD/R-induced injury, which results from activation of the ERK1/2 and p38 MAPK pathways.


Assuntos
Aquaporina 4/fisiologia , Astrócitos/enzimologia , Isquemia Encefálica/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Receptores de Leucotrienos/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Aquaporina 4/biossíntese , Astrócitos/metabolismo , Astrócitos/fisiologia , Western Blotting , Isquemia Encefálica/enzimologia , Isquemia Encefálica/fisiopatologia , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Fosforilação , Ratos , Ratos Sprague-Dawley , Receptores de Leucotrienos/efeitos dos fármacos , Receptores de Leucotrienos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , SRS-A/análogos & derivados , SRS-A/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Regulação para Cima/efeitos dos fármacos
18.
Sheng Li Xue Bao ; 62(2): 101-8, 2010 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-20401443

RESUMO

The aim of the present study is to investigate the role of nordihydroguaiaretic acid (NDGA) on inflammatory cells accumulation after focal cerebral ischemia and the underlying mechanism. Focal cerebral ischemia was induced by 30 min of middle cerebral artery occlusion (MCAO) followed by 72 h of reperfusion. NDGA (5 and 10 mg/kg) was administered intraperitoneally 30 min, 2, 24, 48 h after reperfusion, respectively. The brain injuries were observed by neurological and histological examination. Endogenous IgG exudation, neutrophils and macrophages/microglia accumulation, and intercellular adhesion molecule-1 (ICAM-1) protein expression were determined by immunohistochemistry 72 h after reperfusion. ICAM-1 mRNA was determined by RT-PCR 72 h after reperfusion. The catalysates of 5-lipoxygenase (5-LOX), leukotriene B4 (LTB4) and cysteinyl leukotrienes (CysLTs), were evaluated by ELISA 3 h after reperfusion. The results showed that NDGA ameliorated neurological dysfunction, decreased infarct volume, and inhibited endogenous IgG exudation, neutrophils infiltration, ICAM-1 mRNA and protein expression 72 h after reperfusion. Moreover, NDGA reduced the levels of LTB4 and CysLTs 3 h after reperfusion. However, NDGA did not reduce the accumulation of macrophages/microglia 72 h after reperfusion. These results suggest that NDGA decreases neutrophil infiltration in the subacute phase of focal cerebral ischemia via inhibiting 5-LOX activation.


Assuntos
Araquidonato 5-Lipoxigenase/metabolismo , Isquemia Encefálica/fisiopatologia , Inflamação/fisiopatologia , Inibidores de Lipoxigenase/farmacologia , Masoprocol/farmacologia , Neutrófilos/efeitos dos fármacos , Animais , Isquemia Encefálica/complicações , Imunoglobulina G/imunologia , Inflamação/etiologia , Inflamação/prevenção & controle , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Leucotrieno B4/metabolismo , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/prevenção & controle
19.
Acta Pharmacol Sin ; 31(2): 137-44, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20139896

RESUMO

AIM: To determine whether the flavonoid baicalin attenuates oxygen-glucose deprivation (OGD)-induced injury by inhibiting oxidative stress-mediated 5-lipoxygenase (5-LOX) activation in PC12 cells. METHODS: The effects of baicalin and the 5-LOX inhibitor zileuton on the changes induced by OGD/recovery or H(2)O(2) (an exogenous reactive oxygen species [ROS]) in green fluorescent protein-5-LOX-transfected PC12 cells were compared. RESULTS: Both baicalin and zileuton attenuated OGD/recovery- and H(2)O(2)-induced injury and inhibited OGD/recovery-induced production of 5-LOX metabolites (cysteinyl leukotrienes) in a concentration-dependent manner. However, baicalin did not reduce baseline cysteinyl leukotriene levels. Baicalin also reduced OGD/recovery-induced ROS production and inhibited 5-LOX translocation to the nuclear envelope and p38 phosphorylation induced by OGD/recovery and H(2)O(2). In contrast, zileuton did not show these effects. CONCLUSION: Baicalin can inhibit 5-LOX activation after ischemic injury, which may partly result from inhibition of the ROS/p38 mitogen-activated protein kinase pathway.


Assuntos
Araquidonato 5-Lipoxigenase/metabolismo , Flavonoides/farmacologia , Glucose/metabolismo , Estresse Oxidativo , Oxigênio/metabolismo , Animais , Ativação Enzimática , Peróxido de Hidrogênio/metabolismo , Células PC12 , Fosforilação , Ratos , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Life Sci ; 86(5-6): 170-7, 2010 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-20006627

RESUMO

AIMS: We previously reported that minocycline attenuates acute brain injury and inflammation after focal cerebral ischemia, and this is partly mediated by inhibition of 5-lipoxygenase (5-LOX) expression. Here, we determined the protective effect of minocycline on chronic ischemic brain injury and its relation with the inhibition of 5-LOX expression after focal cerebral ischemia. MAIN METHODS: Focal cerebral ischemia was induced by 90 min of middle cerebral artery occlusion followed by reperfusion for 36 days. Minocycline (45 mg/kg) was administered intraperitoneally 2h and 12h after ischemia and then every 12h for 5 days. Sensorimotor function was evaluated 1-28 days after ischemia and cognitive function was determined 30-35 days after ischemia. Thereafter, infarct volume, neuron density, astrogliosis, and 5-LOX expression in the brain were determined. KEY FINDINGS: Minocycline accelerated the recovery of sensorimotor and cognitive functions, attenuated the loss of neuron density, and inhibited astrogliosis in the boundary zone around the ischemic core, but did not affect infarct volume. Minocycline significantly inhibited the increased 5-LOX expression in the proliferated astrocytes in the boundary zone, and in the macrophages/microglia in the ischemic core. SIGNIFICANCE: Minocycline accelerates functional recovery in the chronic phase of focal cerebral ischemia, which may be partly associated with the reduction of 5-LOX expression.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Inibidores de Lipoxigenase , Minociclina/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Agnosia/etiologia , Agnosia/prevenção & controle , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/enzimologia , Astrócitos/patologia , Encéfalo/enzimologia , Encéfalo/patologia , Isquemia Encefálica/enzimologia , Isquemia Encefálica/fisiopatologia , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Doença Crônica , Imuno-Histoquímica , Injeções Intraperitoneais , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/patologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Microglia/efeitos dos fármacos , Microglia/enzimologia , Microglia/patologia , Minociclina/administração & dosagem , Minociclina/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/patologia , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA