Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 220
Filtrar
1.
Ann Oncol ; 27(1): 185-92, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26487585

RESUMO

BACKGROUND: Anaplastic lymphoma kinase (ALK) fusions need to be accurately and efficiently detected for ALK inhibitor therapy. Fluorescence in situ hybridization (FISH) remains the reference test. Although increasing data are supporting that ALK immunohistochemistry (IHC) is highly concordant with FISH, IHC screening needed to be clinically and prospectively validated. PATIENTS AND METHODS: In the AF-001JP trial for alectinib, 436 patients were screened for ALK fusions through IHC (n = 384) confirmed with FISH (n = 181), multiplex RT-PCR (n = 68), or both (n = 16). IHC results were scored with iScore. RESULT: ALK fusion was positive in 137 patients and negative in 250 patients. Since the presence of cancer cells in the samples for RT-PCR was not confirmed, ALK fusion negativity could not be ascertained in 49 patients. IHC interpreted with iScore showed a 99.4% (173/174) concordance with FISH. All 41 patients who had iScore 3 and were enrolled in phase II showed at least 30% tumor reduction with 92.7% overall response rate. Two IHC-positive patients with an atypical FISH pattern responded to ALK inhibitor therapy. The reduction rate was not correlated with IHC staining intensity. CONCLUSIONS: Our study showed (i) that when sufficiently sensitive and appropriately interpreted, IHC can be a stand-alone diagnostic for ALK inhibitor therapies; (ii) that when atypical FISH patterns are accompanied by IHC positivity, the patients should be considered as candidates for ALK inhibitor therapies, and (iii) that the expression level of ALK fusion is not related to the level of response to ALK inhibitors and is thus not required for patient selection. REGISTRATION NUMBER: JapicCTI-101264 (This study is registered with the Japan Pharmaceutical Information Center).


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/uso terapêutico , Carbazóis/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Proteínas de Fusão Oncogênica/metabolismo , Piperidinas/uso terapêutico , Receptores Proteína Tirosina Quinases/metabolismo , Adenocarcinoma/enzimologia , Adenocarcinoma/mortalidade , Adulto , Idoso , Idoso de 80 Anos ou mais , Quinase do Linfoma Anaplásico , Intervalo Livre de Doença , Feminino , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/mortalidade , Masculino , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Estudos Prospectivos , Resultado do Tratamento , Adulto Jovem
2.
Neuroscience ; 231: 206-15, 2013 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-23238575

RESUMO

Neuronal electrical activity has been known to affect the viability of neurons in the central nervous system. Here we show that long-lasting membrane depolarization induced by elevated extracellular K(+) recruits nitric oxide (NO)/soluble guanylyl cyclase/protein kinase G signaling pathway, induces 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP)-mediated protein S-guanylation, and confers dopaminergic neuroprotection. Treatment of primary mesencephalic cell cultures with 1-methyl-4-phenylpyridinium (MPP(+)) for 72 h decreased the number of dopaminergic neurons, whereas the cell loss was markedly inhibited by elevated extracellular concentration of K(+) (+40 mM). The neuroprotective effect of elevated extracellular K(+) was significantly attenuated by tetrodotoxin (a Na(+) channel blocker), amlodipine (a voltage-dependent Ca(2+) channel blocker), N(ω)-nitro-l-arginine methyl ester (l-NAME) (a nitric oxide synthase inhibitor), 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) (a soluble guanylyl cyclase inhibitor), and KT5823 or Rp-8-bromo-ß-phenyl-1,N(2)-ethenoguanosine 3',5'-cyclic monophosphorothioate (Rp-8-Br-PET-cGMPS) (protein kinase G inhibitors). Elevated extracellular K(+) increased 8-nitro-cGMP production resulting in the induction of protein S-guanylation in cells in mesencephalic cultures including dopaminergic neurons. In addition, exogenous application of 8-nitro-cGMP protected dopaminergic neurons from MPP(+) cytotoxicity, which was prevented by zinc protoporphyrin IX, an inhibitor of heme oxygenase-1 (HO-1). Zinc protoporphyrin IX also inhibited the neuroprotective effect of elevated extracellular K(+). On the other hand, KT5823 or Rp-8-Br-PET-cGMPS did not inhibit the induction of HO-1 protein expression by 8-nitro-cGMP, although these protein kinase G inhibitors abrogated the neuroprotective effect of 8-nitro-cGMP. These results suggest that protein S-guanylation (leading to HO-1 induction) as well as canonical protein kinase G signaling pathway plays an important role in NO-mediated, activity-dependent dopaminergic neuroprotection.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Guanilato Ciclase/metabolismo , Mesencéfalo/metabolismo , Óxido Nítrico/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais/fisiologia , 1-Metil-4-fenilpiridínio/farmacologia , Animais , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Doadores de Óxido Nítrico/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Guanilil Ciclase Solúvel , Tionucleotídeos/farmacologia
3.
Neuroscience ; 222: 10-9, 2012 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-22820264

RESUMO

We have demonstrated previously that nicotine affords neuroprotective and anti-inflammatory effects against intracerebral hemorrhage (ICH)-associated neuropathological changes. The present study was undertaken to clarify whether subtype-specific agonists of nicotinic acetylcholine receptors (nAChRs) could preserve tissue integrity in mouse ICH model in vivo. ICH was induced by unilateral injection of collagenase into the striatum of male C57BL/6 mice. Daily intraperitoneal injection of α7 nAChR agonist PNU-282987 (3-10mg/kg) for 3 days, starting from 3h after induction of ICH, significantly increased the number of surviving neurons in the central and the peripheral regions of hematoma at 3 days after ICH. In contrast, α4ß2 nAChR agonist RJR-2403 (2-10 mg/kg) given in the same regimen showed no significant effect. PNU-282987 and RJR-2403 did not affect either the size of hemorrhage or the extent of brain edema associated with ICH. PNU-282987 decreased the number of activated microglia/macrophages accumulating in the perihematoma region at 3 days after ICH, in a dose-dependent manner. On the other hand, the number of microglia/macrophages in the central region of hematoma at early phase of pathology (6 h after ICH) was increased by 10mg/kg PNU-282987. These results suggest that α7 nAChR agonist can provide neuroprotective effect on ICH-induced injury, independently of its anti-inflammatory actions.


Assuntos
Hemorragia Cerebral/tratamento farmacológico , Hemorragia Cerebral/patologia , Agonistas Nicotínicos/uso terapêutico , Receptores Nicotínicos/efeitos dos fármacos , Animais , Western Blotting , Água Corporal/metabolismo , Encéfalo/patologia , Química Encefálica/efeitos dos fármacos , Edema Encefálico/patologia , Contagem de Células , Hemorragia Cerebral/induzido quimicamente , Colagenases , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/patologia , Neurônios/patologia , Nicotina/análogos & derivados , Nicotina/uso terapêutico , Receptor Nicotínico de Acetilcolina alfa7
4.
Br J Pharmacol ; 166(3): 1151-68, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22224485

RESUMO

BACKGROUND AND PURPOSE: Caffeic acid phenethyl ester (CAPE) is a component of honey bee propolis that can induce expression of haem oxygenase-1 (HO-1). Because HO-1 induction has been suggested to protect dopaminergic neurons in the substantia nigra, we examined the effect of CAPE in experimental models of dopaminergic neurodegeneration. EXPERIMENTAL APPROACH: Neuroprotective effect of CAPE was investigated in rat organotypic midbrain slice cultures and in vivo, using a mouse model of dopaminergic neurodegeneration induced by intranigral injection of LPS and intrastriatal injection of 6-hydroxydopamine. KEY RESULTS: CAPE protected dopaminergic neurons in slice cultures from IFN-γ/LPS-induced injury. The effect of CAPE was inhibited by zinc protoporphyrin IX, an HO-1 inhibitor, and by neutralizing antibody against brain-derived neurotrophic factor (BDNF). A p38 MAPK inhibitor SB203580 prevented activation of NF-E2-related factor 2, attenuated increased expression of HO-1 and BDNF, and blocked the neuroprotective actions of CAPE. In the LPS-injected mouse model, daily intraperitoneal administration of CAPE protected dopaminergic neurons, up-regulated HO-1 and BDNF, and reduced the increase of activated microglia/macrophages. Neuroprotective effects of CAPE against LPS-induced injury was prevented by zinc protoporphyrin IX or anti-BDNF antibody. CAPE protected dopaminergic neurons and alleviated methamphetamine-induced rotational behaviour also in 6-hydroxydopamine hemiparkinsonian mice. CONCLUSION AND IMPLICATIONS: CAPE is a novel type of neuroprotective agent whose actions are mediated by both HO-1 and BDNF. These findings may provide novel clues to develop neuroprotective agents for treatment of neurodegenerative disorders.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Ácidos Cafeicos/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Heme Oxigenase-1/biossíntese , Fármacos Neuroprotetores/farmacologia , Substância Negra/efeitos dos fármacos , Animais , Western Blotting , Ácidos Cafeicos/uso terapêutico , Modelos Animais de Doenças , Neurônios Dopaminérgicos/enzimologia , Neurônios Dopaminérgicos/metabolismo , Indução Enzimática , Heme Oxigenase-1/antagonistas & inibidores , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/uso terapêutico , Óxido Nítrico/metabolismo , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/enzimologia , Transtornos Parkinsonianos/metabolismo , Álcool Feniletílico/análogos & derivados , Ratos , Ratos Wistar , Substância Negra/enzimologia , Substância Negra/metabolismo
5.
Neuroscience ; 190: 289-300, 2011 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-21712074

RESUMO

Narcolepsy results from disruption of orexin neurons in the hypothalamus that play a key role in maintenance of the arousal state. Underlying mechanisms leading to selective loss of orexin neurons remain unknown. On the other hand, endoplasmic reticulum stress, namely, conditions associated with impairment of endoplasmic reticulum functions such as proper folding and sorting of newly synthesized proteins, is implicated in pathogenesis of several types of neurodegenerative disorders. Here we found that application of endoplasmic reticulum stress inducers such as tunicamycin (that prevents protein N-glycosylation) and thapsigargin (that inhibits Ca²âº-ATPase) to organotypic slice cultures of the hypothalamus caused preferential loss of orexin-immunoreactive neurons, as compared to melanin-concentrating hormone- or calcitonin gene-related peptide-immunoreactive neurons. The decrease in orexin-immunoreactive neurons at early time points (6-24 h) was not accompanied by induction of cell death as indicated by the absence of caspase-3 activation and no significant change in the number of NeuN-positive cells, whereas sustained treatment with tunicamycin for 72 h induced cell death. At 24-h treatment, tunicamycin and thapsigargin did not decrease expression of prepro-orexin mRNA, suggesting that post-transcriptional mechanisms were responsible for depletion of orexin peptides. In addition, inhibition of axonal transport by colchicine and inhibition of proteasomal activity by MG132 significantly prevented the decrease in orexin immunoreactivity by tunicamycin. Comparative examinations of expression of unfolded protein response-related proteins revealed that C/EBP-homologous protein (a transcription factor that promotes induction of apoptosis) as well as phosphorylated form of RNA-dependent protein kinase-like endoplasmic reticulum kinase (a protein kinase that mediates inhibition of protein translation) was expressed more prominently in orexin neurons than in melanin-concentrating hormone neurons, in response to tunicamycin. These results indicate that orexin neurons are particularly sensitive to endoplasmic reticulum stress, which may be relevant to pathogenic events in narcolepsy.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Retículo Endoplasmático/metabolismo , Hipotálamo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Contagem de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hormônios Hipotalâmicos/metabolismo , Hipotálamo/patologia , Melaninas/metabolismo , Narcolepsia/metabolismo , Narcolepsia/patologia , Neurônios/patologia , Orexinas , Técnicas de Cultura de Órgãos , Fosforilação , Hormônios Hipofisários/metabolismo , Ratos , Ratos Wistar
6.
Neuroscience ; 170(1): 298-307, 2010 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-20620197

RESUMO

Orexins are neuropeptides produced in the lateral hypothalamus and implicated in regulation of sleep-wake cycle. Selective loss of orexin neurons is found in the brain of patients with narcolepsy, but the mechanisms of this pathological change are unclear. A previous study showed that excessive stimulation of N-methyl-d-aspartate (NMDA) receptors by quinolinic acid (QA) caused selective loss of orexin neurons in rat hypothalamic slice culture. Here we examined QA toxicity on orexin neurons and melanin-concentrating hormone (MCH) neurons in vivo. Contrary to the expectation, injection of QA (60 and 120 nmol) into the lateral hypothalamus of male C57BL/6 mice caused selective loss of MCH neurons rather than orexin neurons, and this toxicity of QA was attenuated by MK-801, an NMDA receptor antagonist. Selective loss of MCH neurons with preserved orexin neurons was observed even when GABA(A) receptor antagonists such as bicuculline and picrotoxin were injected with QA. A significant decrease in the number of orexin neurons was induced when QA injection was performed in the dark phase of diurnal cycle, but the degree of the decrease was still lower than that in the number of MCH neurons. Finally, QA (60 nmol) induced selective loss of MCH neurons also in young rats at 3-4 weeks of age. These results do not support the hypothesis that acute excitotoxicity mediated by NMDA receptors is responsible for the pathogenesis of narcolepsy.


Assuntos
Hormônios Hipotalâmicos/metabolismo , Hipotálamo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Melaninas/metabolismo , Melanóforos/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Hormônios Hipofisários/metabolismo , Ácido Quinolínico/toxicidade , Fatores Etários , Animais , Contagem de Células/métodos , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/fisiologia , Hormônios Hipotalâmicos/antagonistas & inibidores , Hipotálamo/efeitos dos fármacos , Hipotálamo/patologia , Masculino , Melaninas/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/patologia , Orexinas , Hormônios Hipofisários/antagonistas & inibidores , Distribuição Aleatória , Ratos , Ratos Wistar
7.
Neuroscience ; 158(2): 856-66, 2009 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-18996444

RESUMO

Excessive production of nitric oxide (NO) by microglia is at least in part responsible for the pathogenesis of various neurodegenerative disorders including Parkinson disease, but at the same time NO may also play a distinct role as a signaling molecule such as an activator of soluble guanylyl cyclase. Here we investigated potential roles of the NO-soluble guanylyl cyclase-cyclic GMP signaling pathway in the regulation of dopaminergic neurodegeneration. Activation of microglia by interferon-gamma (IFN-gamma) followed by lipopolysaccharide (LPS) caused dopaminergic cell death in rat midbrain slice cultures, which was dependent on NO production. 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ), a soluble guanylyl cyclase inhibitor, as well as KT5823, an inhibitor of cyclic GMP-dependent protein kinase, exacerbated dopaminergic cell death induced by IFN-gamma/LPS. Conversely, 8-bromo-cyclic GMP attenuated IFN-gamma/LPS cytotoxicity on dopaminergic neurons. Notably, although heme oxygenase-1 (HO-1) was expressed prominently in cells other than dopaminergic neurons in control cultures, robust expression of HO-1 was induced in surviving dopaminergic neurons challenged with IFN-gamma/LPS. ODQ and KT5823 decreased, whereas 8-bromo-cyclic GMP increased, the number of dopaminergic neurons expressing HO-1 after IFN-gamma/LPS challenge, without parallel changes in HO-1 expression in other cell populations. An NO donor 3-(4-morpholinyl)sydnonimine hydrochloride also induced HO-1 expression in dopaminergic neurons, which was abolished by ODQ and augmented by 8-bromo-cyclic GMP. Moreover, IFN-gamma/LPS-induced dopaminergic cell death was augmented by zinc protoporphyrin IX, an HO-1 inhibitor. The NO donor cytotoxicity on dopaminergic neurons was also augmented by ODQ and zinc protoporphyrin IX. These results indicate that the NO-cyclic GMP signaling pathway promotes the induction of HO-1 specifically in dopaminergic neurons, which acts as an endogenous protective system to limit inflammatory degeneration of this cell population.


Assuntos
GMP Cíclico/metabolismo , Dopamina/metabolismo , Heme Oxigenase-1/metabolismo , Mesencéfalo/patologia , Degeneração Neural/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais/fisiologia , Animais , Animais Recém-Nascidos , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/genética , Técnicas In Vitro , Inflamação/induzido quimicamente , Inflamação/complicações , Interferon gama , Degeneração Neural/tratamento farmacológico , Degeneração Neural/etiologia , Polissacarídeos , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Tirosina 3-Mono-Oxigenase/metabolismo
8.
Neuroscience ; 144(2): 694-701, 2007 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-17084034

RESUMO

Intracerebral hemorrhage represents stroke characterized by formation and expansion of hematoma within brain parenchyma. Blood-derived factors released from hematoma are considered to be involved in poor prognosis of this disorder. We previously reported that thrombin, a blood-derived serine protease, induced cytotoxicity in the cerebral cortex and the striatum in organotypic slice cultures, which depended on mitogen-activated protein kinase (MAPK) pathways. Here we investigated the mechanisms of thrombin cytotoxicity in the striatum in vivo. Thrombin microinjected into the striatum of adult rats induced neuronal death and microglial activation around the injection site. Neuronal loss without any sign of nuclear fragmentation was observed as early as 4 h after thrombin injection, which was followed by gradual neuronal death exhibiting nuclear fragmentation. Thrombin-induced damage assessed at 72 h after injection was partially but significantly reduced by concomitant administration of inhibitors of MAPK pathways. Activation of extracellular signal-regulated kinase (ERK) and p38 MAPK in response to thrombin was verified by Western blot analysis. Moreover, phosphorylated ERK and p38 MAPK were localized prominently in reactive microglia, and inhibition of microglial activation by minocycline attenuated thrombin-induced damage, suggesting that reactive microglia were responsible for thrombin-induced neuronal death. Thus, MAPK pathways and microglial activation may serve as therapeutic targets of pathogenic conditions associated with hemorrhagic stroke.


Assuntos
Corpo Estriado/patologia , Hemostáticos/toxicidade , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Síndromes Neurotóxicas/etiologia , Transdução de Sinais/fisiologia , Trombina/toxicidade , Animais , Antígeno CD11b/metabolismo , Contagem de Células , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Lateralidade Funcional , Imuno-Histoquímica/métodos , Masculino , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Fosfopiruvato Hidratase/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
9.
J Neural Transm (Vienna) ; 113(2): 151-8, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15959853

RESUMO

Selegiline, a therapeutic agent of Parkinson's disease, is known to have neuroprotective properties that may involve its regulatory effects on antioxidant enzymes. We evaluated effects of selegiline on activities of catalase (CAT), Cu,Zn-superoxide dismutase (SOD1) and Mn-SOD (SOD2) in the striatum, cortex and hippocampus of 8- and 25-week-old rats, and on SOD activities and glutathione levels in mesencephalic slice cultures. Selegiline (2 mg/kg) significantly increased CAT and SOD2 activities in the striatum, but not in the cortex and hippocampus, of 25-week-old rats. In contrast, selegiline failed to increase CAT and SOD activities in three brain regions of 8-week-old rats, whereas L: -dopa significantly increased SOD1 activity in the striatum. In slice cultures, selegiline increased SOD1 and SOD2 activities with a maximal effective concentration of 10(-8) and 10(-10) M, respectively. Moreover, selegiline significantly increased glutathione level. These results suggest that selegiline can decrease oxidative stress in nigrostriatum by augmenting various antioxidant systems, each of which responds optimally to different concentrations of selegiline.


Assuntos
Antioxidantes/metabolismo , Antiparkinsonianos/farmacologia , Corpo Estriado/efeitos dos fármacos , Selegilina/farmacologia , Substância Negra/efeitos dos fármacos , Animais , Catalase/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Corpo Estriado/metabolismo , Ativação Enzimática/efeitos dos fármacos , Glutationa/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Levodopa/farmacologia , Masculino , Técnicas de Cultura de Órgãos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar , Substância Negra/metabolismo , Superóxido Dismutase/metabolismo
10.
Neuroscience ; 116(2): 465-75, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12559101

RESUMO

D-fructose-1,6-bisphosphate, a high energy glycolytic intermediate, attenuates ischemic damage in a variety of tissues, including brain. To determine whether D-fructose-1,6-bisphosphate serves as an alternate energy substrate in the CNS, rat hippocampal slices were treated with D-fructose-1,6-bisphosphate during glucose deprivation. Unlike pyruvate, an endproduct of glycolysis, 10 mM D-fructose-1,6-bisphosphate did not preserve synaptic transmission or morphological integrity of CA1 pyramidal neurons during glucose deprivation. Moreover, during glucose deprivation, 10-mM D-fructose-1,6-bisphosphate failed to maintain adenosine triphosphate levels in slices. D-fructose-1,6-bisphosphate, however, attenuated acute neuronal degeneration produced by 200 microM iodoacetate, an inhibitor of glycolysis downstream of D-fructose-1,6-bisphosphate. Because (5S, 10R)-(+)-5-methyl-10, 11-dihydro-5H-dibenzo [a,d]cyclohepten-5,10-imine, an antagonist of N-methyl-D-aspartate receptors, exhibited similar protection against iodoacetate damage, we examined whether (5S, 10R)-(+)-5-methyl-10, 11-dihydro-5H-dibenzo [a,d]cyclohepten-5,10-imine and D-fructose-1,6-bisphosphate share a common neuroprotective mechanism. Indeed, D-fructose-1,6-bisphosphate diminished N-methyl-D-aspartate receptor-mediated synaptic responses and partially attenuated neuronal degeneration induced by 100-microM N-methyl-D-aspartate. Taken together, these results indicate that D-fructose-1,6-bisphosphate is unlikely to serve as an energy substrate in the hippocampus, and that neuroprotective effects of D-fructose-1,6-bisphosphate are mediated by mechanisms other than anaerobic energy supply.


Assuntos
Metabolismo Energético/efeitos dos fármacos , Frutosedifosfatos/farmacologia , Hipocampo/citologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Glucose/farmacologia , Hipocampo/metabolismo , Neurônios/metabolismo , Técnicas de Cultura de Órgãos , Ácido Pirúvico/farmacologia , Ratos , Receptores de N-Metil-D-Aspartato/fisiologia , Transmissão Sináptica/efeitos dos fármacos
11.
Eur J Pharmacol ; 421(3): 181-9, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11516435

RESUMO

"Catecholaminergic and serotoninergic activity enhancer" effects are newly found mechanisms of action of a class of compound that enhance impulse propagation-mediated release of catecholamines and serotonin in the brain. In the present study, (-)-1-(benzofuran-2-yl)-2-propylaminopentane hydrochloride [(-)-BPAP HCl], a compound with selective and potent "catecholaminergic and serotoninergic activity enhancer" effects, was tested for its efficacy to potentiate locomotor activity in normal rats and to attenuate hypolocomotion in reserpine-treated rats. (-)-BPAP HCl potentiated locomotor activity in non-habituated rats during a 2-h observation period dose-dependently (0.3-10 mg/kg). (-)-BPAP HCl (1-3 mg/kg) was also effective to reverse reserpine-induced hypolocomotion. The effects of (-)-BPAP HCl in normal and reserpine-treated rats were attenuated by the dopamine D1 receptor antagonist, R(+)-7-chloro-8-hydroxy-3-methyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine (SCH 23390), suggesting that the effects of (-)-BPAP HCl were mediated by activation of the dopaminergic system. In addition, the administration of (-)-BPAP HCl increased ipsilateral turning in unilaterally 6-hydroxydopamine-lesioned rats, implying presynaptic activation of nigrostriatal dopaminergic terminals by (-)-BPAP HCl. Furthermore, although antiparkinsonian agents, such as apomorphine and amantadine, failed to improve reserpine-induced ptosis, (-)-BPAP HCl significantly improved ptosis. These findings suggested that a "catecholaminergic and serotoninergic activity enhancer" compound, (-)-BPAP, stimulates motor function in rats and improves motor deficits in animal models of Parkinson's disease due to its ability to induce dopamine release.


Assuntos
Benzofuranos/farmacologia , Dopamina/metabolismo , Atividade Motora/efeitos dos fármacos , Amantadina/farmacologia , Animais , Antipsicóticos/farmacologia , Benzazepinas/farmacologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Dopaminérgicos/farmacologia , Antagonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Habituação Psicofisiológica , Masculino , Oxidopamina/farmacologia , Ratos , Ratos Wistar , Reserpina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo
12.
J Chromatogr B Biomed Sci Appl ; 757(1): 127-33, 2001 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-11419737

RESUMO

In this study, a simple, sensitive and enantioselective HPLC method was developed for the simultaneous determination of lansoprazole enantiomers: a proton pump inhibitor, and its major metabolites: 5-hydroxylansoprazole and lansoprazole sulfone in human liver microsomes. After extraction from the microsomal incubation mixture with a diethyl etherdichloromethane (7:3, v/v) mixture, analytes were measured by reversed-phase HPLC on a Chiralcel OD-R column. Detection was made using an ultraviolet absorbance detector set at a wavelength of 285 nm. The mobile phase consisted of a methanol-water (75:25, v/v) mixture. At a flow-rate of 0.5 ml/min, the total run time was 35 min. The limit of quantification for both lansoprazole enantiomers was 0.25 microM and for the metabolites 0.13 microM. The method is suitable for the analysis of lansoprazole enantiomers and its metabolites from human microsomal liver incubations.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Microssomos Hepáticos/metabolismo , Omeprazol/análogos & derivados , Omeprazol/metabolismo , 2-Piridinilmetilsulfinilbenzimidazóis , Calibragem , Humanos , Lansoprazol , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Espectrofotometria Ultravioleta , Estereoisomerismo
13.
Neuropharmacology ; 40(6): 761-71, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11369030

RESUMO

This study was undertaken to determine whether 1 alpha,25-dihydroxyvitamin D3 [1 alpha,25-(OH)(2)D(3)], an active metabolite of vitamin D, protects dopaminergic neurons against the neurotoxic effects of glutamate and dopaminergic toxins using rat mesecephalic culture. Brief glutamate exposure elicited cytotoxicity in both dopaminergic and non-dopaminergic neurons. Pretreatment, but not co-administration, of 1 alpha,25-(OH)(2)D(3) protected both types of neurons against the cytotoxicity of glutamate in a concentration- and time-dependent manner. The neuroprotective effect of 1 alpha,25-(OH)(2)D(3) was inhibited by the protein synthesis inhibitor, cycloheximide. To investigate the mechanisms of these neuroprotective effects, we examined the effects of 1 alpha,25-(OH)(2)D(3) on neurotoxicity induced by calcium ionophore and reactive oxygen species (ROS). Pretreatment with 1 alpha,25-(OH)(2)D(3) protected both types of neurons against the cytotoxicity induced by A23187 in a concentration-dependent manner. Furthermore, 24-h pretreatment with 1 alpha,25-(OH)(2)D(3) concentration-dependently protected both types of neurons from ROS-induced cytotoxicity. A 24-h incubation with 1 alpha,25-(OH)(2)D(3) inhibited the increase in intracellular ROS level following H(2)O(2) exposure. A 24-h exposure to 1-methyl-4-phenylpyridium ion (MPP(+)) or 6-hydroxydopamine (6-OHDA) exerted selective neurotoxicity on dopaminergic neurons, and these neurotoxic effects were ameliorated by 1 alpha,25-(OH)(2)D(3). These results suggest that 1 alpha,25-(OH)(2)D(3) provides protection of dopaminergic neurons against cytotoxicity induced by glutamate and dopaminergic toxins by facilitating cellular functions that reduce oxidative stress.


Assuntos
Ácido Glutâmico/toxicidade , Mesencéfalo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Vitamina D/farmacologia , 1-Metil-4-fenilpiridínio/toxicidade , Adrenérgicos/toxicidade , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Dopamina/metabolismo , Herbicidas/toxicidade , Mesencéfalo/metabolismo , Neurônios/fisiologia , Oxidopamina/toxicidade , Ratos , Receptores de Calcitriol/metabolismo , Vitamina D/análogos & derivados
14.
J Neurochem ; 76(5): 1336-45, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11238718

RESUMO

We have previously demonstrated that dopaminergic neurons in midbrain-striatum slice co-cultures are more resistant to NMDA cytotoxicity than the same neuronal population in single midbrain slice cultures. Here, we show that dopaminergic neurons in midbrain-striatum co-cultures also exhibit resistance to the cytotoxicity of nitric oxide donors, 2,2'-(hydroxynitrosohydrazono)bis-ethanamine (NOC-18) and 3-morpholinosydnonimine (SIN-1). The cytotoxicity of NMDA (30 microM) in single cultures was significantly attenuated by the nitric oxide synthase (NOS) inhibitor N(omega)-nitro-L-arginine (100 microM), whereas the toxicity in co-cultures was not. The levels of tyrosine residue nitration of tyrosine hydroxylase, a hallmark of the occurence of peroxynitrite anion in dopaminergic neurons, were lower in co-cultures than those in single cultures. Single cultures and co-cultures did not show appreciable differences in the number or distribution of NOS-containing neurons as assessed by NADPH diaphorase histochemistry. On the other hand, midbrain slices cultured with striatal slices showed higher levels of superoxide dismutase (SOD) activity as well as increased protein levels of Cu,Zn-SOD, than midbrain slices cultured alone. These results suggested that the generation of NO is involved in NMDA cytotoxicity on dopaminergic neurons, and that increased activity of SOD in co-cultures renders dopaminergic neurons resistant to NMDA cytotoxicity by preventing the formation of peroxynitrite.


Assuntos
Corpo Estriado/fisiologia , Dopamina/metabolismo , Mesencéfalo/fisiologia , N-Metilaspartato/farmacologia , Neurônios/fisiologia , Neurotoxinas/farmacologia , Doadores de Óxido Nítrico/farmacologia , Superóxido Dismutase/metabolismo , Animais , Animais Recém-Nascidos , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Di-Hidrolipoamida Desidrogenase/metabolismo , Molsidomina/análogos & derivados , Molsidomina/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Nitroarginina/farmacologia , Compostos Nitrosos/farmacologia , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Tirosina/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
15.
Neurosci Lett ; 300(3): 166-70, 2001 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-11226637

RESUMO

Chronic treatment of organotypic midbrain slice cultures with L-type Ca(2+) channel blocker nicardipine (3-10 microM) or verapamil (10 microM) for 18 days resulted in a drastic decrease in the number of dopaminergic neurons. A voltage-dependent Na(+) channel blocker tetrodotoxin (1 microM) was also effective in decreasing the number of dopaminergic neurons. Concurrent application of forskolin (20 microM) or dibutyryl cyclic AMP (1 mM) counteracted the effects of nicardipine and tetrodotoxin. These results suggest that spontaneous neuronal activity within midbrain slice cultures, causing Ca(2+) influx through L-type Ca(2+) channels that maintains intracellular cyclic AMP levels, is required for the maintenance of dopaminergic neurons.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Dopamina/metabolismo , Mesencéfalo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Nicardipino/farmacologia , Verapamil/farmacologia , Animais , Animais Recém-Nascidos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Colforsina/farmacologia , Técnicas de Cultura , Mesencéfalo/fisiologia , Neurônios/fisiologia , Ratos , Ratos Wistar , Tetrodotoxina/farmacologia
16.
Eur J Clin Pharmacol ; 57(10): 709-15, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11829200

RESUMO

OBJECTIVE: The aim of this investigation was to clarify the stereoselective properties in lansoprazole metabolism by monitoring the metabolic consumption for each enantiomer and the formation of the main metabolites, lansoprazole sulfone and 5-hydroxylansoprazole, in the presence of human liver microsomal enzymes. METHODS: Human liver microsomes or recombinant cytochrome P450 (CYP) enzymes were incubated with either (+/- )-, (+)-, or (-)-lansoprazole in the presence of reduced nicotinamide adenine dinucleotide phosphate. The metabolic consumption of lansoprazole enantiomers was estimated from the amounts of enantiomers consumed by microsomal enzymes after incubation at 37 degrees C for 60 min. Metabolites of lansoprazole, lansoprazole sulfone, and 5-hydroxylansoprazole were determined after incubation at 37 degrees C for 20 min, and kinetic parameters [Michaelis constant (Km) and maximum velocity (Vmax)] were obtained using Eadie-Hofstee plots. RESULTS: (-)-Lansoprazole was metabolized more preferentially than (+)-lansoprazole in human liver microsomes. Stereoselective sulfoxidation and hydroxylation [(+) > (-)] were observed in human liver microsomes. Strikingly, in sulfoxidation, a significantly higher intrinsic clearance (Vmax,l/Km,l) of (-)-lansoprazole (0.023 +/- 0.001 ml/min/mg) than (+)-lansoprazole (0.006 +/- 0.000 ml/min/mg) was observed. Consequently, sulfoxidation is likely to play an important role in the stereoselective metabolism of lansoprazole enantiomers. P450-isoform specificity for each enantiomer was evident. CYP3A4, which mainly catalyzed sulfoxidation, was more active toward (-)-lansoprazole in either a chiral or racemic drug as a substrate. CYP2C19, which catalyzed hydroxylation, preferentially metabolized (+)-lansoprazole. The consumption of (+)-lansoprazole was markedly inhibited by (-)-lansoprazole, indicating a metabolic enantiomer/enantiomer interaction. However, this alteration of recombinant CYP2C19 specificity for (+)-lansoprazole did not appear in metabolism in human liver microsomes. CONCLUSIONS: Stereoselective metabolism was observed in human liver microsomes, and this stereoselectivity was mainly based on CYP3A4 specificity for preferable metabolism of (-)-lansoprazole.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/fisiologia , Inibidores Enzimáticos/metabolismo , Microssomos Hepáticos/metabolismo , Oxigenases de Função Mista/fisiologia , Omeprazol/análogos & derivados , Omeprazol/metabolismo , 2-Piridinilmetilsulfinilbenzimidazóis , Biotransformação , Citocromo P-450 CYP2C19 , Citocromo P-450 CYP3A , Inibidores Enzimáticos/farmacocinética , Humanos , Hidroxilação , Técnicas In Vitro , Lansoprazol , Omeprazol/farmacocinética , Isoformas de Proteínas , Inibidores da Bomba de Prótons , Estereoisomerismo
17.
Brain Res ; 885(2): 240-50, 2000 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-11102578

RESUMO

We investigated possible involvement of voltage-dependent Ca(2+) channels (VDCCs) and several intracellular signaling mechanisms in multiple actions of basic fibroblast growth factor (bFGF), such as survival promotion, induction of calbindin D(28k) expression as well as acceleration of neuritic branch formation of cultured rat hippocampal neurons. Immunocytochemical staining with anti-gamma-aminobutyric acid (GABA) antibody showed that the promotion of neuron survival by bFGF in high cell-density cultures were exerted exclusively on GABA-negative neurons. Nicardipine (5 microM) attenuated the effect of bFGF on neuronal survival and formation of neurite branches, suggesting that the activity of L-type VDCCs is required for these effects. In contrast, stimulation of calbindin expression by bFGF was not attenuated by nicardipine. A phospholipase C inhibitor U73122 (1 microM) prevented the effect of bFGF on neurite branch formation, but not on neuronal survival or calbindin expression. On the other hand, chronic application of phorbol-12-myristate-13-acetate (1 microM) inhibited the effect of bFGF on neuronal survival, without inhibiting the other bFGF actions. Forskolin (100 microM) attenuated the effect of bFGF on neuronal survival and neurite branch formation, indicating that cyclic AMP plays negative regulatory roles in these actions of bFGF. Taken together, these results suggest that multiple biological actions of bFGF on hippocampal neurons are exerted through, and modulated by, distinct signaling pathways.


Assuntos
Canais de Cálcio Tipo L/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Neuritos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Proteína G de Ligação ao Cálcio S100/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Calbindinas , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Embrião de Mamíferos , Hipocampo/citologia , Neuritos/metabolismo , Neurônios/metabolismo , Nicardipino/farmacologia , Ratos , Ratos Wistar , Proteína G de Ligação ao Cálcio S100/metabolismo , Transdução de Sinais/fisiologia
18.
Phys Rev Lett ; 84(15): 3286-9, 2000 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-11019071

RESUMO

Rotation-vibration transitions of methane molecules embedded in parahydrogen crystals were investigated through Fourier transform infrared spectroscopy. Each transition shows extremely sharp peaks with a Lorentzian line shape profile, which indicates the spectra are free from inhomogeneous broadening. The steep temperature dependence of the linewidths observed in the range between 3.7 and 8. 5 K is interpreted to be a result of the pure dephasing relaxation mechanism. A remarkable difference in population relaxation widths between stretching and bending vibrational excited states was also found.

19.
Anesth Analg ; 91(4): 944-8, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11004054

RESUMO

UNLABELLED: Highly concentrated lidocaine has been reported to induce irreversible loss of membrane potential in crayfish nerve, which implies membrane disruption as one of the direct mechanisms of lidocaine-induced neurotoxicity. To confirm lidocaine-induced membrane disruption in mammalian nerve, a lactate dehydrogenase (LDH) leakage from rat sciatic nerve was measured in vitro. Before applying lidocaine, the desheathed nerve was incubated for 60 min in Krebs-Ringer solution at 37 degrees C to examine basal LDH activity. It was then incubated in 80 mM lidocaine solution at pH 7.3 for 15, 30, 60, or 120 min. Other nerves were immersed in 800 mM choline solution for 120 min. Total LDH activity per wet weight of nerve tissue was assayed using spectrophotometry. It was also determined using nerves cut into 10 segments and incubated in distilled water for 60 min. The LDH activity in the lidocaine group showed a time-dependent increase. After the 60- and 120-min incubation with lidocaine, the amount of LDH activity was significantly increased compared with the choline group and was similar to that of the group incubated in distilled water. We conclude that 80 mM lidocaine may be sufficient to cause membrane damage and facilitate the leakage of enzymes from cytoplasm. IMPLICATIONS: This study demonstrates that exposing the rat myelinated nerve to lidocaine at a clinically used concentration for more than 30 min causes enough membrane damage to allow enzyme leakage. In clinical practice, the smallest effective dose should be used.


Assuntos
Anestésicos Locais/farmacologia , Axônios/efeitos dos fármacos , Lidocaína/farmacologia , Nervo Isquiático/efeitos dos fármacos , Análise de Variância , Animais , Axônios/enzimologia , Morte Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/enzimologia , Colina/farmacologia , L-Lactato Desidrogenase/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Nootrópicos/farmacologia , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/enzimologia , Espectrofotometria , Fatores de Tempo
20.
Eur J Pharmacol ; 404(1-2): 41-8, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10980261

RESUMO

This study was performed to examine the roles of the N-methyl-D-aspartate (NMDA) receptor/phencyclidine (PCP) channel complex in the protective effects of sigma-receptor ligands against glutamate neurotoxicity in cultured cortical neurons derived from fetal rats. A 1-h exposure of cultures to glutamate caused a marked loss of viability, as determined by Trypan blue exclusion. This acute neurotoxicity of glutamate was prevented by NMDA receptor antagonists. Expression of sigma(1) receptor mRNA in cortical cultures was confirmed by reverse transcription polymerase chain reaction (RT-PCR). sigma Receptor ligands with affinity for NMDA receptor channels including the PCP site, such as (+)-N-allylnormetazocine ((+)-SKF10,047), haloperidol, and R(-)-N-(3-phenyl-1-propyl)-1-phenyl-2-aminopropane ((-)-PPAP), prevented glutamate neurotoxicity in a concentration-dependent manner. In contrast, other sigma-receptor ligands without affinity for NMDA receptors, such as carbetapentane and R(+)-3-(3-hydroxyphenyl)-N-propylpiperidine ((+)-3-PPP), did not show neuroprotective effects. Putative endogenous sigma receptor ligands such as pregnenolone, progesterone, and dehydroepiandrosterone did not affect glutamate neurotoxicity. The protective effects of (+)-SKF10,047, haloperidol, and (-)-PPAP were not affected by the sigma(1) receptor antagonist rimcazole. These results suggested that a direct interaction with NMDA receptors but not with sigma receptors plays a crucial role in the neuroprotective effects of sigma receptor ligands with affinity for NMDA receptors.


Assuntos
Ácido Glutâmico/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores Opioides delta/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Haloperidol/farmacologia , Ligantes , Neurônios/metabolismo , Fenazocina/análogos & derivados , Fenazocina/farmacologia , Propilaminas/farmacologia , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...