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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 27(35): 9278-93, 2007 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-17728442

RESUMO

Loss of mitochondrial membrane integrity and release of apoptogenic factors are a key step in the signaling cascade leading to neuronal cell death in various neurological disorders, including ischemic injury. Emerging evidence has suggested that the intramitochondrial protein apoptosis-inducing factor (AIF) translocates to the nucleus and promotes caspase-independent cell death induced by glutamate toxicity, oxidative stress, hypoxia, or ischemia. However, the mechanism by which AIF is released from mitochondria after neuronal injury is not fully understood. In this study, we identified calpain I as a direct activator of AIF release in neuronal cultures challenged with oxygen-glucose deprivation and in the rat model of transient global ischemia. Normally residing in both neuronal cytosol and mitochondrial intermembrane space, calpain I was found to be activated in neurons after ischemia and to cleave intramitochondrial AIF near its N terminus. The truncation of AIF by calpain activity appeared to be essential for its translocation from mitochondria to the nucleus, because neuronal transfection of the mutant AIF resistant to calpain cleavage was not released after oxygen-glucose deprivation. Adeno-associated virus-mediated overexpression of calpastatin, a specific calpain-inhibitory protein, or small interfering RNA-mediated knockdown of calpain I expression in neurons prevented ischemia-induced AIF translocation. Moreover, overexpression of calpastatin or knockdown of AIF expression conferred neuroprotection against cell death in neuronal cultures and in hippocampal CA1 neurons after transient global ischemia. Together, these results define calpain I-dependent AIF release as a novel signaling pathway that mediates neuronal cell death after cerebral ischemia.


Assuntos
Fator de Indução de Apoptose/metabolismo , Calpaína/metabolismo , Hipóxia/metabolismo , Mitocôndrias/metabolismo , Neurônios/patologia , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Calpaína/genética , Células Cultivadas , Modelos Animais de Doenças , Eletroforese em Gel de Campo Pulsado/métodos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Glucose/deficiência , Humanos , Hipóxia/fisiopatologia , Marcação In Situ das Extremidades Cortadas/métodos , Proteínas Mitocondriais/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Transfecção/métodos
2.
J Neurochem ; 106(2): 860-74, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18466318

RESUMO

To study whether and how cells adapt to chronic cellular stress, we exposed PC12 cells to the proteasome inhibitor MG132 (0.1 microM) for 2 weeks and longer. This treatment reduced chymotrypsin-like proteasome activity by 47% and was associated with protection against both 6-hydroxydopamine (6-OHDA; 100 microM) and higher dose MG132 (40 microM). Protection developed slowly over the course of the first 2 weeks of exposure and was chronic thereafter. There was no change in total GSH levels after MG132. Buthionine sulfoximine (100 microM) reduced GSH levels by 60%, but exacerbated 6-OHDA toxicity to the same extent in both MG132-treated and control cells and failed to reduce MG132-induced protection. Chronic MG132 resulted in elevated antioxidant proteins CuZn superoxide dismutase (SOD; +55%), MnSOD (+21%), and catalase (+15%), as well as chaperone heat-shock protein 70 (+42%). Examination of SOD enzyme activity revealed higher levels of CuZnSOD (+40%), with no change in MnSOD. We further assessed the mechanism of protection by reducing CuZnSOD levels with two independent siRNA sequences, both of which successfully attenuated protection against 6-OHDA. Previous reports suggested that artificial over-expression of CuZnSOD in dopaminergic cells is protective. Our data complement such observations, revealing that dopaminergic cells are also able to use endogenous CuZnSOD in self-defensive adaptations to chronic stress, and that they can even do so in the face of extensive GSH loss.


Assuntos
Adaptação Biológica/efeitos dos fármacos , Inibidores de Cisteína Proteinase/farmacologia , Leupeptinas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Superóxido Dismutase/fisiologia , Adrenérgicos/farmacologia , Animais , Butionina Sulfoximina/farmacologia , Catalase/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Dopamina/metabolismo , Inibidores Enzimáticos/farmacologia , Glutationa/metabolismo , Oxidopamina/farmacologia , Células PC12/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , RNA Interferente Pequeno/farmacologia , Ratos , Tirosina 3-Mono-Oxigenase/metabolismo
3.
Neurobiol Dis ; 32(1): 116-24, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18675914

RESUMO

Leucine-rich repeat kinase 2 (LRRK2) is a recently identified gene that, when mutated at specific locations, results in the onset of parkinsonian symptoms with clinical features indistinguishable from idiopathic Parkinson's disease. Based on structural and domain analysis, LRRK2 is predicted to function as a stress-responsive protein scaffold mediating the regulation of mitogen activating protein kinase (MAPK) pathways. Consistent with this notion, our results supported the notion that expression of wild-type LRRK2 but not Y1699C or G2019S mutants enhanced the tolerance of HEK293 and SH-SY5Y cells towards H(2)O(2)-induced oxidative stress. This increase in stress tolerance was dependent on the presence of the kinase domain of the LRRK2 gene and manifested through the activation of the ERK pathway. Collectively, our results indicated that cells expressing LRRK2 mutants suffer a loss of protection normally derived from wild-type LRRK2, making them more vulnerable to oxidative stress.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Mutação/fisiologia , Estresse Oxidativo/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Transdução de Sinais/fisiologia , Morte Celular/genética , Morte Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Sistema de Sinalização das MAP Quinases/genética , Mutação/genética , Estresse Oxidativo/genética , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/genética
4.
Cell Transplant ; 26(7): 1224-1234, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28933217

RESUMO

Scaffolds composed of extracellular matrix (ECM) are being investigated for their ability to facilitate brain tissue remodeling and repair following injury. The present study tested the hypothesis that the implantation of brain-derived ECM would attenuate experimental traumatic brain injury (TBI) and explored potential underlying mechanisms. TBI was induced in mice by a controlled cortical impact (CCI). ECM was isolated from normal porcine brain tissue by decellularization methods, prepared as a hydrogel, and injected into the ipsilesional corpus callosum and striatum 1 h after CCI. Lesion volume and neurological function were evaluated up to 35 d after TBI. Immunohistochemistry was performed to assess post-TBI white matter integrity, reactive astrogliosis, and microglial activation. We found that ECM treatment reduced lesion volume and improved neurobehavioral function. ECM-treated mice showed less post-TBI neurodegeneration in the hippocampus and less white matter injury than control, vehicle-treated mice. Furthermore, ECM ameliorated TBI-induced gliosis and microglial pro-inflammatory responses, thereby providing a favorable microenvironment for tissue repair. Our study indicates that brain ECM hydrogel implantation improved the brain microenvironment that facilitates post-TBI tissue recovery. Brain ECM offers excellent biocompatibility and holds potential as a therapeutic agent for TBI, alone or in combination with other treatments.


Assuntos
Lesões Encefálicas Traumáticas/fisiopatologia , Lesões Encefálicas Traumáticas/terapia , Encéfalo/metabolismo , Matriz Extracelular/transplante , Recuperação de Função Fisiológica , Animais , Comportamento Animal , Lesões Encefálicas Traumáticas/patologia , Região CA3 Hipocampal/patologia , Região CA3 Hipocampal/fisiopatologia , Gliose/patologia , Implantes Experimentais , Inflamação/patologia , Masculino , Camundongos Endogâmicos C57BL , Neuroglia/patologia , Neurônios/patologia , Fármacos Neuroprotetores/uso terapêutico , Substância Branca/patologia
5.
FASEB J ; 19(10): 1350-2, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15941767

RESUMO

This study attempted to elucidate the signaling mechanism underlying dopaminergic cell death in the MPP+ model for Parkinson's disease. In neuronal-differentiated PC12 cells, through the regulation by activated JNK and c-jun, BimEL expression was markedly increased in response to MPP+ treatment, which led to the cell degeneration. In lieu of Smac translocation as seen in other paradigms, up-regulation of BimEL effected an increase in calpain I activity that, in turn, mediated AIF release from the mitochondria. In support, we found that knocking down BimEL expression resulted in a decrease in calpain I activity, as well as AIF release from the mitochondria and cell death. Finally, inhibition of calpain activity mitigated AIF release from the mitochondria and cell death. Under cell-free conditions, activated purified calpain I could induce the release of AIF from isolated mitochondria without the participation of BimEL or activated JNK, suggesting that AIF release is a direct consequence of calpain I activity. In concert, the results suggest a novel signaling pathway for dopaminergic cell degeneration, in which MPP+ induces the up-regulation of BimEL, which in turn potentiates an elevation in calpain I activity that mediates AIF release and cell death in a caspase-independent manner.


Assuntos
1-Metil-4-fenilpiridínio/toxicidade , Fator de Indução de Apoptose/metabolismo , Proteínas Reguladoras de Apoptose/fisiologia , Morte Celular/efeitos dos fármacos , Proteínas de Membrana/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Animais , Proteína 11 Semelhante a Bcl-2 , Cálcio/metabolismo , Calpaína/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Células PC12 , Transporte Proteico/efeitos dos fármacos , Ratos , Regulação para Cima
6.
Prog Neurobiol ; 69(2): 103-42, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12684068

RESUMO

After a severe episode of ischemia, traumatic brain injury (TBI) or epilepsy, it is typical to find necrotic cell death within the injury core. In addition, a substantial number of neurons in regions surrounding the injury core have been observed to die via the programmed cell death (PCD) pathways due to secondary effects derived from the various types of insults. Apart from the cell loss in the injury core, cell death in regions surrounding the injury core may also contribute to significant losses in neurological functions. In fact, it is the injured neurons in these regions around the injury core that treatments are targeting to preserve. In this review, we present our cumulated understanding of stress-activated signaling pathways and apoptotic pathways in the research areas of ischemic injury, TBI and epilepsy and that gathered from concerted research efforts in oncology and other diseases. However, it is obvious that our understanding of these pathways in the context of acute brain injury is at its infancy stage and merits further investigation. Hopefully, this added research effort will provide a more detailed knowledge from which better therapeutic strategies can be developed to treat these acute brain injuries.


Assuntos
Apoptose/fisiologia , Lesões Encefálicas/patologia , Epilepsia/patologia , Isquemia/patologia , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Animais , Lesões Encefálicas/fisiopatologia , Epilepsia/fisiopatologia , Humanos , Isquemia/fisiopatologia , Neurônios/metabolismo , Estresse Fisiológico/patologia , Estresse Fisiológico/fisiopatologia
7.
FASEB J ; 17(3): 520-2, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12551850

RESUMO

Our results presented here suggest that cortical neurons degenerate via two caspase-mediated apoptotic pathways when challenged with 0.5 microM rotenone. Although these two pathways can be attributed to the loss of mitochondrial integrity, the triggers for these pathways are likely due to two separate subsequent events (the release of cytochrome c and the emergence of mitochondrial permeability transition [MPT]). Despite some reports suggesting that the release of cytochrome c is a consequence of MPT, the results of our time course experiments suggest otherwise. In fact, we observed that the release of cytochrome c occurred much earlier than MPT; hence, the former is unlikely to be a consequence of the latter. In addition, we observed that the presence of an MPT inhibitor did not attenuate the activation of caspase-3, and that caspase-1-mediated cell death did not exhibit nuclear condensation and DNA fragmentation. Taken together, these results suggest that there are two routes by which cortical neurons degenerate during ischemic injury or in neurodegenerative diseases.


Assuntos
Apoptose , Caspases/fisiologia , Córtex Cerebral/citologia , Neurônios/enzimologia , Rotenona/toxicidade , Animais , Caspase 1/metabolismo , Caspase 3 , Caspases/metabolismo , Células Cultivadas , Grupo dos Citocromos c/metabolismo , Canais Iônicos/metabolismo , Proteínas de Transporte da Membrana Mitocondrial , Poro de Transição de Permeabilidade Mitocondrial , Modelos Biológicos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Estresse Oxidativo , Transdução de Sinais
8.
Sci Rep ; 5: 9621, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25858671

RESUMO

Galectin-1 (gal-1), a special lectin with high affinity to ß-galactosides, is implicated in protection against ischemic brain injury. The present study investigated transplantation of gal-1-secreting neural stem cell (s-NSC) into ischemic brains and identified the mechanisms underlying protection. To accomplish this goal, secretory gal-1 was stably overexpressed in NE-4C neural stem cells. Transient cerebral ischemia was induced in mice by middle cerebral artery occlusion for 60 minutes and s-NSCs were injected into the striatum and cortex within 2 hours post-ischemia. Brain infarct volume and neurological performance were assessed up to 28 days post-ischemia. s-NSC transplantation reduced infarct volume, improved sensorimotor and cognitive functions, and provided more robust neuroprotection than non-engineered NSCs or gal-1-overexpressing (but non-secreting) NSCs. White matter injury was also ameliorated in s-NSC-treated stroke mice. Gal-1 modulated microglial function in vitro, by attenuating secretion of pro-inflammatory cytokines (TNF-α and nitric oxide) in response to LPS stimulation and enhancing production of anti-inflammatory cytokines (IL-10 and TGF-ß). Gal-1 also shifted microglia/macrophage polarization toward the beneficial M2 phenotype in vivo by reducing CD16 expression and increasing CD206 expression. In sum, s-NSC transplantation confers robust neuroprotection against cerebral ischemia, probably by alleviating white matter injury and modulating microglial/macrophage function.


Assuntos
Isquemia Encefálica/metabolismo , Galectina 1/metabolismo , Células-Tronco Neurais/metabolismo , Transplante de Células-Tronco , Animais , Comportamento Animal , Isquemia Encefálica/imunologia , Isquemia Encefálica/patologia , Isquemia Encefálica/terapia , Sobrevivência Celular , Infarto Cerebral/patologia , Infarto Cerebral/terapia , Corpo Caloso/patologia , Corpo Estriado/patologia , Citocinas/biossíntese , Macrófagos/metabolismo , Camundongos , Microglia/metabolismo , Óxido Nítrico/metabolismo , Fagocitose , Fenótipo , Desempenho Psicomotor , Fatores de Tempo , Substância Branca/patologia
9.
Sci Rep ; 5: 10224, 2015 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-25959573

RESUMO

Major characteristics of Alzheimer's disease (AD) include deposits of ß-amyloid (Aß) peptide in the brain, loss of synapses, and cognitive dysfunction. Cocaine- and amphetamine-regulated transcript (CART) has recently been reported to attenuate Aß-induced toxicity. In this study, CART localization in APP/PS1 mice was characterized and the protective effects of exogenous CART treatment were examined. Compared to age-matched wild type mice, 8-month-old APP/PS1 mice had significantly greater CART immunoreactivity in the hippocampus and cortex. A strikingly similar pattern of Aß plaque-associated CART immunoreactivity was observed in the cortex of AD cases. Treatment of APP/PS1 mice with exogenous CART ameliorated memory deficits; this effect was associated with improvements in synaptic ultrastructure and long-term potentiation, but not a reduction of the Aß plaques. Exogenous CART treatment in APP/PS1 mice prevented depolarization of the mitochondrial membrane and stimulated mitochondrial complex I and II activities, resulting in an increase in ATP levels. CART treatment of APP/PS1 mice also reduced reactive oxygen species and 4-hydroxynonenal, and mitigated oxidative DNA damage. In summary, CART treatment reduced multiple neuropathological measures and improved memory in APP/PS1 mice, and may therefore be a promising and novel therapy for AD.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Memória/efeitos dos fármacos , Proteínas do Tecido Nervoso/administração & dosagem , Neurotransmissores/administração & dosagem , Sinapses/efeitos dos fármacos , Sinapses/patologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Estudos de Casos e Controles , Dano ao DNA , DNA Mitocondrial , Modelos Animais de Doenças , Expressão Gênica , Hipocampo/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neurotransmissores/genética , Neurotransmissores/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Placa Amiloide/patologia
10.
Prog Neurobiol ; 119-120: 60-84, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24923657

RESUMO

Microglia are the first line of immune defense against central nervous system (CNS) injuries and disorders. These highly plastic cells play dualistic roles in neuronal injury and recovery and are known for their ability to assume diverse phenotypes. A broad range of surface receptors are expressed on microglia and mediate microglial 'On' or 'Off' responses to signals from other host cells as well as invading microorganisms. The integrated actions of these receptors result in tightly regulated biological functions, including cell mobility, phagocytosis, the induction of acquired immunity, and trophic factor/inflammatory mediator release. Over the last few years, significant advances have been made toward deciphering the signaling mechanisms related to these receptors and their specific cellular functions. In this review, we describe the current state of knowledge of the surface receptors involved in microglial activation, with an emphasis on their engagement of distinct functional programs and their roles in CNS injuries. It will become evident from this review that microglial homeostasis is carefully maintained by multiple counterbalanced strategies, including, but not limited to, 'On' and 'Off' receptor signaling. Specific regulation of theses microglial receptors may be a promising therapeutic strategy against CNS injuries.


Assuntos
Sistema Nervoso Central/imunologia , Microglia/fisiologia , Traumatismos do Sistema Nervoso/imunologia , Animais , Humanos , Receptores de Superfície Celular/metabolismo , Receptores de Quimiocinas/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Receptores Purinérgicos/metabolismo
11.
J Neurochem ; 99(4): 1151-63, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16956375

RESUMO

Exposure to sublethal stress can trigger endogenous protection against subsequent, higher levels of stress. We tested for this preconditioning phenomenon in a model of Parkinson's disease by applying 6-hydroxydopamine to the dopaminergic MN9D cell line. Exposure to sublethal concentrations of 6-hydroxydopamine (5-10 microM) protected against the toxic effects of a subsequent exposure to a higher concentration (50 microM), as measured by the Hoechst assay for nuclear viability. This was accompanied by little or no protection against 6-hydroxydopamine-induced lactate dehydrogenase release, decline in ATP, or reduction in (3)H-dopamine uptake. The antioxidant, N-acetyl cysteine (20 mM), when applied during preconditioning, abolished protection, as did the protein synthesis inhibitor, cycloheximide (0.2 microM). Preconditioning did not affect superoxide dismutase or glutathione peroxidase enzymes, or levels of heat shock protein-72. However, Bcl-2 protein levels rose with preconditioning. Preconditioning rapidly increased phosphorylation of kinases ERK1/2, Akt and JNK, and was abolished by pharmacological inhibitors of their activity. Finally, sublethal 6-hydroxydopamine preconditioned against the toxicity of proteasome inhibitor, MG-132 (1 microM). Thus, exposure of a dopaminergic cell line to sublethal oxidative stress can protect against additional oxidative stress due to translational and post-translational modifications, as well as confer 'cross-tolerance' against a different insult, proteasome inhibition.


Assuntos
Citoproteção/fisiologia , Dopamina/metabolismo , Resistência a Medicamentos/fisiologia , Neurônios/metabolismo , Estresse Oxidativo/fisiologia , Substância Negra/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Antioxidantes/farmacologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular , Citoproteção/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Oxidopamina/toxicidade , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/fisiologia , Inibidores da Síntese de Proteínas/farmacologia , Substância Negra/efeitos dos fármacos , Substância Negra/fisiopatologia , Simpatolíticos/toxicidade
12.
J Neurosci Res ; 77(5): 747-61, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15352222

RESUMO

This study showed that primary dopaminergic neurons or the dopaminergic cell line MN9D, when exposed to 15 min of the parkinsonian toxin 6-hydroxydopamine (6-OHDA) in the range of 30-100 microM, underwent delayed degeneration and exhibited hallmarks of apoptosis. These results, along with the absence of any increase in lactate dehydrogenase (LDH) release from the degenerated cells, imply that apoptosis was the dominant mode of cell death. Moreover, a distinct elevation in the measured cellular activities of caspase-9 and -3 but not of caspase-8 points to the caspase-9/caspase-3 cascade as the predominant apoptotic pathway in the degeneration of dopaminergic neurons and MN9D cells. In addition, the presence of caspase-9 or -3 peptide inhibitors but not of caspase-8 inhibitor attenuated cell death significantly, supporting the notion that only the intrinsic apoptotic pathway is utilized to achieve cell death. Finally, overexpression of a mutant caspase-9 with dominant negative phenotype (caspase-9dn) in MN9D cells and primary dopaminergic neurons via the adenovirus and adenoassociated virus gene delivery system, respectively, conferred marked increases in tolerance to the toxicity of 6-OHDA. These results point to the intrinsic caspase-9/caspase-3 cascade as the predominant signaling pathway underlying dopaminergic cell death induced by 6-OHDA and suggest that gene delivery of caspase-9dn can attenuate this pathway and its degenerative consequences.


Assuntos
Apoptose , Caspases/fisiologia , Dopamina/metabolismo , Degeneração Neural/induzido quimicamente , Oxidopamina/toxicidade , Animais , Animais Recém-Nascidos , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/genética , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Inibidores de Cisteína Proteinase/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Imunofluorescência/métodos , Regulação da Expressão Gênica/fisiologia , L-Lactato Desidrogenase/análise , Mutação , Degeneração Neural/prevenção & controle , Oligopeptídeos/farmacologia , Ratos , Sais de Tetrazólio , Fatores de Tempo , Transfecção/métodos , Tirosina 3-Mono-Oxigenase/metabolismo , Ácido gama-Aminobutírico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA