Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Skin Res Technol ; 30(7): e13845, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39031933

RESUMO

BACKGROUND: Observational studies have shown an association between skin microbiota and alopecia areata (AA), but the causal connection remains ambiguous. METHODS: We obtained data on skin microbiota and AA from summary statistics of Genome-Wide Association Studies and applied statistical methods from Mendelian randomization (MR) to assess causal relationships. Additionally, we investigated whether the skin microbiota acts as a mediator in the pathway from gut microbiota to AA. RESULTS: In the MR analysis of KORA FF4 and AA, the inverse-variance weighting method indicated that Corynebacterium (odds ratio [OR] = 0.82, 95% confidence interval [CI]: 0.70-0.96, p = 0.02) and asv037 (OR = 0.87, 95% CI: 0.76-0.99, p = 0.05) exerted protective effects, while Betaproteobacteria (OR = 1.21, 95% CI: 1.01-1.44, p = 0.03), asv015 (OR = 1.27, 95% CI: 1.05-1.54, p = 0.02), and Burkholderiales (OR = 1.20, 95% CI: 1.04-1.38, p = 0.01) were identified as risk factors in AA. In the MR analysis of PopGen and AA, asv001 (OR = 1.12, 95% CI: 1.01-1.24, p = 0.04), asv054 (OR = 1.13, 95% CI: 1.01-1.25, p = 0.03), and asv059 (OR = 1.14, 95% CI: 1.02-1.27, p = 0.02) were found to potentially increase the risk in AA. Furthermore, in the influence of gut microbiota on AA, the skin microbiota did not act as a mediator. CONCLUSION: Our analysis suggests potential causal relationships between certain skin microbiota and AA, revealing insights into its pathogenesis and potential intervention strategies.


Assuntos
Alopecia em Áreas , Microbioma Gastrointestinal , Análise da Randomização Mendeliana , Pele , Humanos , Alopecia em Áreas/microbiologia , Alopecia em Áreas/genética , Microbioma Gastrointestinal/fisiologia , Pele/microbiologia , Estudo de Associação Genômica Ampla , Microbiota/genética
2.
Brain Inj ; 22(10): 723-32, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18720098

RESUMO

PRIMARY OBJECTIVE: Recent efforts have been aimed at developing a panel of protein biomarkers for the diagnosis/prognosis of the neurological damage associated with acute brain injury. METHODS AND PROCEDURES: This study utilized high-throughput immunoblotting (HTPI) technology to compare changes between two animal models of acute brain injury: penetrating ballistic-like brain injury (PBBI) which mimics the injury created by a gunshot wound and transient middle cerebral artery occlusion (MCAo) which is a model of stroke. Brain and blood were collected at 24-hours post-injury. MAIN OUTCOMES AND RESULTS: This study identified the changes in 18 proteins following PBBI and 17 proteins following MCAo out of a total of 998 screened proteins. Distinct differences were observed between the two models: five proteins were up- or down-regulated in both models, 23 proteins changed in only one model and one protein was differentially expressed. Western blots were used to verify HTPI results for selected proteins with measurable changes observed in both blood and brain for the proteins STAT3, Tau, PKA RII beta, 14-3-3 epsilon and p43/EMAPII. CONCLUSIONS: These results suggest distinct post-injury protein profiles between brain injury types (traumatic vs. ischemic) that will facilitate strategies aimed at the differential diagnosis and prognosis of acute brain injury.


Assuntos
Hemorragia Cerebral Traumática/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Proteínas de Membrana/análise , Ferimentos por Arma de Fogo/metabolismo , Animais , Biomarcadores/análise , Western Blotting , Química Encefálica , Hemorragia Cerebral Traumática/patologia , Immunoblotting/métodos , Infarto da Artéria Cerebral Média/patologia , Masculino , Modelos Animais , Ratos , Ratos Sprague-Dawley , Ferimentos por Arma de Fogo/patologia
3.
Neurosci Lett ; 418(2): 165-9, 2007 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-17398001

RESUMO

Cellular injury can involve the aberrant stimulation of cell cycle proteins in part through activation of phosphodiesterases (PDEs) and downstream expression of cell-cycle components such as cyclin D1. In mature non-proliferating cells activation of the cell cycle can lead to the induction of programmed cell death. In the present study, we investigated the in vitro neuroprotective efficacy and mechanism of action of vinpocetine (PDE1 inhibitor), trequinsin (PDE3 inhibitor), and rolipram (PDE4 inhibitor) in four mechanistically-distinct models of injury to primary rat cortical neurons as related to cell cycle regulation and apoptosis. Cellular injury was induced by hypoxia/hypoglycemia, veratridine (10 microM), staurosporine (1 microM), or glutamate (100 microM), resulting in average neuronal cell death rates of 43-48% as determined by MTT assay. Treatment with each PDE inhibitor (PDEI) resulted in a similar concentration-dependent neuroprotection profile with maximal effective concentrations of 5-10 microM (55-77% neuroprotection) in all four neurotoxicity models. Direct cytotoxicity due to PDE inhibition alone was not observed at concentrations below 100 microM. Further studies indicated that PDEIs can suppress the excitotoxic upregulation of cyclin D1 similar to the effects of flavopiridol, a cyclin-dependent kinase inhibitor, including suppression of pro-apoptotic caspase-3 activity. Overall, these data indicate that PDEIs are broad-spectrum neuroprotective agents acting through modulation of cell cycle elements and may offer a novel mode of therapy against acute injury to the brain.


Assuntos
Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Proteínas de Ciclo Celular/efeitos dos fármacos , Degeneração Neural/tratamento farmacológico , Neurônios/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Animais , Apoptose/fisiologia , Caspase 3/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/enzimologia , Córtex Cerebral/fisiopatologia , Ciclina D1/efeitos dos fármacos , Ciclina D1/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Degeneração Neural/enzimologia , Degeneração Neural/fisiopatologia , Neurônios/enzimologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Neurotoxinas/antagonistas & inibidores , Neurotoxinas/metabolismo , Inibidores de Fosfodiesterase/uso terapêutico , Diester Fosfórico Hidrolases/efeitos dos fármacos , Diester Fosfórico Hidrolases/metabolismo , Ratos , Ratos Sprague-Dawley
4.
Neurosci Lett ; 422(1): 64-7, 2007 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-17600621

RESUMO

In an earlier study, we demonstrated that PAN-811 (3-aminopyridine-2-carboxaldehyde thiosemicarbazone), a novel neuroprotectant, provides protection against glutamate, staurosporine, veratridine, or hypoxia/hypoglycemia toxicities in primary cortical neuronal cultures by upregulating Bcl-2 expression [R.-W. Chen, C. Yao, X.C. Lu, Z.-G. Jiang, R. Whipple, Z. Liao, H.A. Ghanbari, B. Almassian, F.C. Tortella, J.R. Dave. PAN-811 (3-aminopyridine-2-carboxaldehyde thiosemicarbazone), a novel neuroprotectant, elicits its function in primary neuronal cultures by upregulating Bcl-2 expression. Neuroscience 135 (2005) 191-201]. Both JNK (c-Jun N-terminal kinase) and p38 MAP (mitogen-activated protein) kinase activation have a direct inhibitory action on Bcl-2 by phosphorylation. In the present study, we continued to explore the mechanism of PAN-811 neuroprotection. Our results indicate that treatment of cultured cortical neurons with glutamate (100 microM) induces phosphorylation of both JNK and p38 MAPK. Specifically, pretreatment of neurons with 10 microM PAN-811 (an optimal neuroprotective concentration) for 1h, 4h, or 24h significantly suppresses glutamate-mediated activation of both JNK and p38 MAPK. Furthermore, the p38 MAPK-specific inhibitor SB203580 and the JNK-specific inhibitor SP600125 prevented glutamate-induced neuronal death in these primary cultures. Our results demonstrate that glutamate-induced phosphorylation of JNK and p38 MAPK is suppressed by PAN-811, which might contribute to Bcl-2 upregulation and PAN-811 neuroprotection.


Assuntos
Antagonistas de Aminoácidos Excitatórios , Ácido Glutâmico/toxicidade , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Piridinas/farmacologia , Tiossemicarbazonas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Animais , Antracenos/farmacologia , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Feminino , Genes bcl-2/genética , Gravidez , Ratos , Ratos Sprague-Dawley , Sais de Tetrazólio , Tiazóis
5.
J Neurotrauma ; 26(1): 141-54, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19119917

RESUMO

Glycine-proline-glutamate (GPE) is an N-terminal tripeptide endogenously cleaved from insulin-like growth factor-1 in the brain and is neuroprotective against hypoxic-ischemic brain injury and neurodegeneration. NNZ-2566 is an analog of GPE designed to have improved bioavailability. In this study, we tested NNZ-2566 in a rat model of penetrating ballistic-type brain injury (PBBI) and assessed its effects on injury-induced histopathology, behavioral deficits, and molecular and cellular events associated with inflammation and apoptosis. In the initial dose-response experiments, NNZ-2566 (0.01-3 mg/kg/h x 12 h intravenous infusion) was given at 30 min post-injury and the therapeutic time window was established by delaying treatments 2-4 h post-injury, but with the addition of a 10- or 30-mg/kg bolus dose. All animals survived 72 h. Neuroprotection was evaluated by balance beam testing and histopathology. The effects of NNZ-2566 on injury-induced changes in Bax and Bcl-2 proteins, activated microgliosis, neutrophil infiltration, and astrocyte reactivity were also examined. Behavioral results demonstrated that NNZ-2566 dose-dependently reduced foot faults by 19-66% after acute treatments, and 35-55% after delayed treatments. Although gross lesion volume was not affected, NNZ-2566 treatment significantly attenuated neutrophil infiltration and reduced the number of activated microglial cells in the peri-lesion regions of the PBBI. PBBI induced a significant upregulation in Bax expression (36%) and a concomitant downregulation in Bcl-2 expression (33%), both of which were significantly reversed by NNZ-2566. Collectively, these results demonstrated that NNZ-2566 treatment promoted functional recovery following PBBI, an effect related to the modulation of injury-induced neural inflammatory and apoptotic mechanisms.


Assuntos
Apoptose/efeitos dos fármacos , Lesões Encefálicas/tratamento farmacológico , Encefalite/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Oligopeptídeos/farmacologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/fisiopatologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Encefalite/metabolismo , Encefalite/fisiopatologia , Gliose/tratamento farmacológico , Gliose/fisiopatologia , Gliose/prevenção & controle , Injeções Intravenosas , Microglia/efeitos dos fármacos , Microglia/patologia , Transtornos dos Movimentos/tratamento farmacológico , Transtornos dos Movimentos/etiologia , Transtornos dos Movimentos/fisiopatologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/fisiopatologia , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Oligopeptídeos/agonistas , Oligopeptídeos/química , Oligopeptídeos/uso terapêutico , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/fisiologia , Resultado do Tratamento
6.
J Neurosci Res ; 85(6): 1295-309, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17385714

RESUMO

Cell cycle reentry has been found during apoptosis of postmitotic neurons under certain pathological conditions. To evaluate whether nuclear factor-kappaB (NF-kappaB) activation promotes cell cycle entry and neuronal apoptosis, we studied the relation among NF-kappaB-mediated cyclin induction, bromodeoxyuridine (BrdU) incorporation, and apoptosis initiation in rat striatal neurons following excitotoxic insult. Intrastriatally injected N-methyl-D-aspartate receptor agonist quinolinic acid (QA, 60 nmol) elicited a rise in cyclin D1 mRNA and protein levels (P<0.05). QA-induced NF-kappaB activation occurred in striatal neurons and nonneuronal cells and partially colocalized with elevated cyclin D1 immunoreactivity and TUNEL-positive nuclei. QA triggered DNA replication as evidenced by BrdU incorporation; some striatal BrdU-positive cells were identified as neurons by colocalization with NeuN. Blockade of NF-kappaB nuclear translocation with the recombinant peptide NF-kappaB SN50 attenuated the QA-induced elevation in cyclin D1 and BrdU incorporation. QA-induced internucleosomal DNA fragmentation was blunted by G(1)/S-phase cell cycle inhibitors. These findings suggest that NF-kappaB activation stimulates cyclin D1 expression and triggers DNA replication in striatal neurons. Excitotoxin-induced neuronal apoptosis may thus result from, at least partially, a failed cell cycle attempt.


Assuntos
Apoptose/fisiologia , Corpo Estriado/citologia , Ciclina D1/metabolismo , Replicação do DNA/fisiologia , NF-kappa B/metabolismo , Neurônios/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Análise de Variância , Animais , Apoptose/efeitos dos fármacos , Bromodesoxiuridina/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Replicação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Interações Medicamentosas , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Lateralidade Funcional , Marcação In Situ das Extremidades Cortadas/métodos , Neurônios/efeitos dos fármacos , Peptídeos/farmacologia , Fosfopiruvato Hidratase/metabolismo , Ácido Quinolínico/farmacologia , Ratos , Ratos Sprague-Dawley
7.
J Pharmacol Exp Ther ; 318(3): 947-55, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16728590

RESUMO

Acute seizures following brain injury have been associated with a worsening of patient outcome, but they are often undiagnosed and untreated when they occur without motor convulsions. Here, we sought to compare the antiseizure profile of ethosuximide (EXM; 125-312.5 mg/kg i.v.) and gabapentin (GBP; 0.3-50 mg/kg. i.v.) in a rat model of nonconvulsive seizures (NCS) induced by brain ischemia. Seizures were detected by continuous electroencephalographic monitoring for 24 h following permanent middle cerebral artery occlusion (MCAo). Both "preseizure" and "postseizure" treatment effects were evaluated. Control rats experienced a 91% incidence of NCS (averaging 10-11 NCS/rat), which was significantly reduced following preseizure treatment (delivered 20 min post-MCAo) with either EXM (ED(50) = 161 mg/kg) or GBP (ED(50) = 10.5 mg/kg). In contrast to preseizure treatment effects, only GBP reduced NCS when given after the first seizure event. A further, albeit nonsignificant, 20% reduction in NCS incidence was measured when given in combination postseizure. Drug treatment also reduced infarct volume, which was positively correlated to the number of NCS events (r = 0.475; P < 0.001). EXM and GBP treatment of cultured neurons exposed to neurotoxic or ischemic insults showed no neuroprotective effects, suggesting that in vivo neuroprotection can be attributed to anti-seizure effects. We conclude that EXM and GBP significantly attenuate NCS in a dose-related manner and may help to improve patient outcome from brain ischemia-induced seizure activity.


Assuntos
Aminas/uso terapêutico , Isquemia Encefálica/complicações , Ácidos Cicloexanocarboxílicos/uso terapêutico , Etossuximida/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Convulsões/tratamento farmacológico , Ácido gama-Aminobutírico/uso terapêutico , Doença Aguda , Animais , Relação Dose-Resposta a Droga , Eletroencefalografia , Gabapentina , Masculino , Ratos , Ratos Sprague-Dawley , Convulsões/fisiopatologia
8.
Proc Natl Acad Sci U S A ; 103(5): 1581-6, 2006 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-16423893

RESUMO

Cellular and molecular pathways underlying ischemic neurotoxicity are multifaceted and complex. Although many potentially neuroprotective agents have been investigated, the simplicity of their protective mechanisms has often resulted in insufficient clinical utility. We describe a previously uncharacterized class of potent neuroprotective compounds, represented by PAN-811, that effectively block both ischemic and hypoxic neurotoxicity. PAN-811 disrupts neurotoxic pathways by at least two modes of action. It causes a reduction of intracellular-free calcium as well as free radical scavenging resulting in a significant decrease in necrotic and apoptotic cell death. In a rat model of ischemic stroke, administration of PAN-811 i.c.v. 1 h after middle cerebral artery occlusion resulted in a 59% reduction in the volume of infarction. Human trials of PAN-811 for an unrelated indication have established a favorable safety and pharmacodynamic profile within the dose range required for neuroprotection warranting its clinical trial as a neuroprotective drug.


Assuntos
Infarto da Artéria Cerebral Média/tratamento farmacológico , Isquemia , Doenças Neurodegenerativas/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Piridinas/farmacologia , Tiossemicarbazonas/farmacologia , Animais , Apoptose , Western Blotting , Cálcio/metabolismo , Córtex Cerebral/metabolismo , Quelantes/farmacologia , Técnicas de Cocultura , Corpo Estriado/metabolismo , Fragmentação do DNA , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fluoresceínas/farmacologia , Sequestradores de Radicais Livres/metabolismo , Humanos , Hipóxia , L-Lactato Desidrogenase/metabolismo , Masculino , Necrose , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio , Fatores de Tempo
9.
Proc Natl Acad Sci U S A ; 100(10): 6210-5, 2003 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-12732732

RESUMO

Lithium has long been a primary drug used to treat bipolar mood disorder, even though the drug's therapeutic mechanisms remain obscure. Recent studies demonstrate that lithium has neuroprotective effects against glutamate-induced excitotoxicity in cultured neurons and in vivo. The present study was undertaken to examine whether postinsult treatment with lithium reduces brain damage induced by cerebral ischemia. We found that s.c. injection of lithium dose dependently (0.5-3 mEq/kg) reduced infarct volume in the rat model of middle cerebral artery occlusionreperfusion. Infarct volume was reduced at a therapeutic dose of 1 mEq/kg even when administered up to 3 h after the onset of ischemia. Neurological deficits induced by ischemia were also reduced by daily administration of lithium over 1 week. Moreover, lithium treatment decreased the number of neurons showing DNA damage in the ischemic brain. These neuroprotective effects were associated with an up-regulation of cytoprotective heat shock protein 70 (HSP70) in the ischemic brain hemisphere as determined by immunohistochemistry and Western blotting analysis. Lithium-induced HSP70 up-regulation in the ischemic hemisphere was preceded by an increase in the DNA binding activity of heat shock factor 1, which regulates the transcription of HSP70. Physical variables and cerebral blood flow were unchanged by lithium treatment. Our results suggest that postinsult lithium treatment reduces both ischemia-induced brain damage and associated neurological deficits. Moreover, the heat shock response is likely to be involved in lithium's neuroprotective actions. Additionally, our studies indicate that lithium may have clinical utility for the treatment of patients with acute stroke.


Assuntos
Dano Encefálico Crônico/prevenção & controle , Infarto Cerebral/prevenção & controle , Ataque Isquêmico Transitório/fisiopatologia , Lítio/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Temperatura Corporal/efeitos dos fármacos , Infarto Cerebral/patologia , Circulação Cerebrovascular/efeitos dos fármacos , Ataque Isquêmico Transitório/tratamento farmacológico , Masculino , Ratos , Ratos Sprague-Dawley , Reflexo de Sobressalto , Reperfusão
10.
J Neurochem ; 84(3): 566-75, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12558976

RESUMO

In rat cerebellar granule cells, glutamate induced rapid activation of c-Jun N-terminal kinase (JNK) and p38 kinase to phosphorylate c-Jun (at Ser63) and p53 (at Ser15), respectively, and a subsequent marked increase in activator protein-1 (AP-1) binding that preceded apoptotic death. These glutamate-induced effects and apoptosis could largely be prevented by long-term (7 days) pretreatment with 0.5-2 mm lithium, an antibipolar drug. Glutamate's actions could also be prevented by known blockers of this pathway, MK-801 (an NMDA receptor blocker), SB 203580 (a p38 kinase inhibitor) and curcumin (an AP-1 binding inhibitor). The concentration- and time-dependent suppression of glutamate's effects by lithium and curcumin correlated well with their neuroprotective effects. These results suggest a prominent role of JNK and p38, as well as their downstream AP-1 binding activation and p53 phosphorylation in mediating glutamate excitotoxicity. Moreover, the neuroprotective effects of lithium are mediated, at least in part, by suppressing NMDA receptor-mediated activation of the mitogen-activated protein kinase pathway.


Assuntos
DNA/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neurônios/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Células Cultivadas , Cerebelo/citologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/toxicidade , Proteínas Quinases JNK Ativadas por Mitógeno , Lítio/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Neurônios/citologia , Fármacos Neuroprotetores/farmacologia , Fosforilação/efeitos dos fármacos , Ligação Proteica/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Tempo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
11.
Bipolar Disord ; 4(2): 129-36, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12071510

RESUMO

Lithium, the major drug used to treat manic depressive illness, robustly protects cultured rat brain neurons from glutamate excitotoxicity mediated by N-methyl-D-aspartate (NMDA) receptors. The lithium neuroprotection against glutamate excitotoxiciy is long-lasting, requires long-term pretreatment and occurs at therapeutic concentrations of this drug. The neuroprotective mcchanisms involve inactivation of NMDA receptors, decreased expression of pro-apoptotic proteins, p53 and Bax, enhanced expression of the cytoprotective protein, Bcl-2, and activation of the cell survival kinase, Akt. In addition, lithium pretreatment suppresses glutamate-induced loss of the activities of Akt, cyclic AMP-response element binding protein (CREB), c-Jun - N-terminal kinase (JNK) and p38 kinase. Lithium also reduces brain damage in animal models of neurodegenerative diseases in which excitotoxicity has been implicated. In the rat model of stroke using middle cerebral artery occlusion, lithium markedly reduces neurologic deficits and decreases brain infarct volume even when administered after the onset of ischemia. In a rat Huntington's disease model, lithium significantly reduces brain lesions resulting from intrastriatal infusion of quinolinic acid, an excitotoxin. Our results suggest that lithium might have utility in the treatment of neurodegenerative disorders in addition to its common use for the treatment of bipolar depressive patients.


Assuntos
Carbonato de Lítio/farmacologia , Animais , Transtorno Bipolar/tratamento farmacológico , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Modelos Animais de Doenças , Ácido Glutâmico/metabolismo , Neurônios/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Acidente Vascular Cerebral/tratamento farmacológico
12.
J Neurochem ; 90(5): 1036-49, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15312159

RESUMO

Cyclo-oxygenase-2 (COX-2) is believed to induce neuronal oxidative stress via production of radicals. While oxygen radicals are not directly involved in COX-2-catalytic cycle, superoxide anion radicals have been repeatedly reported to play a critical role in COX-2-associated oxidative stress. To resolve the controversy, we characterized production of free radicals in PC12 cells in which COX-2 expression was manipulated either genetically or by direct protein transfection and compared them with those generated by a recombinant COX-2 in a cell-free system. Using spin-traps alpha-(4-pyridyl-1-oxide)-N-t-butylnitrone, 5,5-dimethyl-1-pyrroline-N-oxide and 4-((9-acridinecarbonyl) amino)-2,2,6,6- tetramethylpiperidine-1-oxyl (Ac-Tempo), we observed arachidonic acid (AA)-dependent production of carbon-centered radicals by heme-reconstituted recombinant COX-2. No oxygen radicals or thiyl radicals have been detected. COX-2 also catalyzed AA-dependent one-electron co-oxidation of ascorbate to ascorbate radicals. Next, we used two different approaches of COX-2 expression in cells, PCXII cells which express isopropyl-1-thio-beta-D-galactopyranoside inducible COX-2, and PC12 cells transfected with COX-2 using a protein delivery reagent, Chariot. In both models, COX-2-dependent AA-induced generation of carbon-centered radicals was documented using spin-traps and Ac-Tempo. No oxygen radical formation was detected in COX-2-transfected cells by either spin-traps or fluorogenic probe, dihydroethidium. In the presence of ascorbate, AA-induced COX-2-dependent ascorbate radicals were detected. AA caused a significant and selective oxidation of one of the major phospholipids, phosphatidylserine (PS). PS was not a direct substrate for COX-2 but was co-oxidized in the presence of AA. The radical generation and PS oxidation were inhibited by COX-2 inhibitors, niflumic acid, nimesulide, or NS-398. Thus, COX-2 generated carbon-centered radicals but not oxygen radicals or thiyl radicals are responsible for oxidative stress in AA-challenged PC12 cells overexpressing COX-2.


Assuntos
Ácido Araquidônico/farmacologia , Carbono/metabolismo , Etídio/análogos & derivados , Radicais Livres/metabolismo , Isoenzimas/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Células PC12/efeitos dos fármacos , Prostaglandina-Endoperóxido Sintases/metabolismo , Animais , Western Blotting/métodos , Cromatografia Líquida de Alta Pressão/métodos , Óxidos N-Cíclicos , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/metabolismo , Interações Medicamentosas , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Etídio/metabolismo , Glicerídeos/farmacologia , Humanos , Peróxido de Hidrogênio/farmacologia , Isoenzimas/genética , Isopropiltiogalactosídeo/farmacologia , Peróxidos Lipídicos/metabolismo , Proteínas de Membrana , Óxidos de Nitrogênio/farmacologia , Oxirredução , Fosfatidilcolinas/farmacologia , Prostaglandina-Endoperóxido Sintases/genética , Piridinas , Ratos , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA