Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Neurosci ; 38(5): 1085-1099, 2018 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-29246925

RESUMO

Dendritic spine loss is recognized as an early feature of Alzheimer's disease (AD), but the underlying mechanisms are poorly understood. Dendritic spine structure is defined by filamentous actin (F-actin) and we observed depolymerization of synaptosomal F-actin accompanied by increased globular-actin (G-actin) at as early as 1 month of age in a mouse model of AD (APPswe/PS1ΔE9, male mice). This led to recall deficit after contextual fear conditioning (cFC) at 2 months of age in APPswe/PS1ΔE9 male mice, which could be reversed by the actin-polymerizing agent jasplakinolide. Further, the F-actin-depolymerizing agent latrunculin induced recall deficit after cFC in WT mice, indicating the importance of maintaining F-/G-actin equilibrium for optimal behavioral response. Using direct stochastic optical reconstruction microscopy (dSTORM), we show that F-actin depolymerization in spines leads to a breakdown of the nano-organization of outwardly radiating F-actin rods in cortical neurons from APPswe/PS1ΔE9 mice. Our results demonstrate that synaptic dysfunction seen as F-actin disassembly occurs very early, before onset of pathological hallmarks in AD mice, and contributes to behavioral dysfunction, indicating that depolymerization of F-actin is causal and not consequent to decreased spine density. Further, we observed decreased synaptosomal F-actin levels in postmortem brain from mild cognitive impairment and AD patients compared with subjects with normal cognition. F-actin decrease correlated inversely with increasing AD pathology (Braak score, Aß load, and tangle density) and directly with performance in episodic and working memory tasks, suggesting its role in human disease pathogenesis and progression.SIGNIFICANCE STATEMENT Synaptic dysfunction underlies cognitive deficits in Alzheimer's disease (AD). The cytoskeletal protein actin plays a critical role in maintaining structure and function of synapses. Using cultured neurons and an AD mouse model, we show for the first time that filamentous actin (F-actin) is lost selectively from synapses early in the disease process, long before the onset of classical AD pathology. We also demonstrate that loss of synaptic F-actin contributes directly to memory deficits. Loss of synaptosomal F-actin in human postmortem tissue correlates directly with decreased performance in memory test and inversely with AD pathology. Our data highlight that synaptic cytoarchitectural changes occur early in AD and they may be targeted for the development of therapeutics.


Assuntos
Actinas/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/fisiologia , Transtornos Cognitivos/genética , Transtornos Cognitivos/psicologia , Espinhas Dendríticas/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Envelhecimento/metabolismo , Doença de Alzheimer/patologia , Animais , Autopsia , Disfunção Cognitiva/patologia , Condicionamento Clássico , Medo/psicologia , Feminino , Humanos , Masculino , Rememoração Mental , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Cultura Primária de Células , Sinaptossomos/metabolismo
2.
Transl Psychiatry ; 13(1): 123, 2023 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-37045867

RESUMO

Women carry a higher burden of Alzheimer's disease (AD) compared to men, which is not accounted entirely by differences in lifespan. To identify the mechanisms underlying this effect, we investigated sex-specific differences in the progression of familial AD in humans and in APPswe/PS1ΔE9 mice. Activity dependent protein translation and associative learning and memory deficits were examined in APPswe/PS1ΔE9 mice and wild-type mice. As a human comparator group, progression of cognitive dysfunction was assessed in mutation carriers and non-carriers from DIAN (Dominantly Inherited Alzheimer Network) cohort. Female APPswe/PS1ΔE9 mice did not show recall deficits after contextual fear conditioning until 8 months of age. Further, activity dependent protein translation and Akt1-mTOR signaling at the synapse were impaired in male but not in female mice until 8 months of age. Ovariectomized APPswe/PS1ΔE9 mice displayed recall deficits at 4 months of age and these were sustained until 8 months of age. Moreover, activity dependent protein translation was also impaired in 4 months old ovariectomized APPswe/PS1ΔE9 mice compared with sham female APPswe/PS1ΔE9 mice. Progression of memory impairment differed between men and women in the DIAN cohort as analyzed using linear mixed effects model, wherein men showed steeper cognitive decline irrespective of the age of entry in the study, while women showed significantly greater performance and slower decline in immediate recall (LOGIMEM) and delayed recall (MEMUNITS) than men. However, when the performance of men and women in several cognitive tasks (such as Wechsler's logical memory) are compared with the estimated year from expected symptom onset (EYO) we found no significant differences between men and women. We conclude that in familial AD patients and mouse models, females are protected, and the onset of disease is delayed as long as estrogen levels are intact.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Humanos , Feminino , Masculino , Camundongos , Animais , Lactente , Doença de Alzheimer/metabolismo , Camundongos Transgênicos , Caracteres Sexuais , Disfunção Cognitiva/genética , Medo , Transtornos da Memória , Modelos Animais de Doenças , Precursor de Proteína beta-Amiloide/genética , Peptídeos beta-Amiloides/metabolismo
3.
Sci Rep ; 11(1): 870, 2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33441593

RESUMO

In this study we demonstrate that 2 month old APPswe/PS1dE9 mice, a transgenic model of Alzheimer's disease, exhibited intact short-term memory in Pavlovian hippocampal-dependent contextual fear learning task. However, their long-term memory was impaired. Intra-CA1 infusion of isoproterenol hydrochloride, the ß-adrenoceptor agonist, to the ventral hippocampus of APPswe/PS1dE9 mice immediately before fear conditioning restored long-term contextual fear memory. Infusion of the ß-adrenoceptor agonist + 2.5 h after fear conditioning only partially rescued the fear memory, whereas infusion at + 12 h post conditioning did not interfere with long-term memory persistence in this mouse model. Furthermore, Intra-CA1 infusion of propranolol, the ß-adrenoceptor antagonist, administered immediately before conditioning to their wildtype counterpart impaired long-term fear memory, while it was ineffective when administered + 4 h and + 12 h post conditioning. Our results indicate that, long-term fear memory persistence is determined by a unique ß-adrenoceptor sensitive time window between 0 and + 2.5 h upon learning acquisition, in the ventral hippocampal CA1 of APPswe/PS1dE9 mice. On the contrary, ß-adrenoceptor agonist delivery to ventral hippocampal CA1 per se did not enhance innate anxiety behaviour in open field test. Thus we conclude that, activation of learning dependent early ß-adrenoceptor modulation underlies and is necessary to promote long-term fear memory persistence in APPswe/PS1dE9.


Assuntos
Medo/fisiologia , Memória/fisiologia , Receptores Adrenérgicos beta/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/fisiologia , Modelos Animais de Doenças , Hipocampo/fisiologia , Masculino , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória , Memória de Longo Prazo/fisiologia , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Transgênicos , Presenilina-1/genética , Presenilina-1/metabolismo
4.
Front Neurosci ; 14: 568200, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33384577

RESUMO

Mild behavioral deficits, which are part of normal aging, can be early indicators of an impending Alzheimer's disease. Using the APPswe/PS1dE9 (APP/PS1) mouse model of Alzheimer's disease, we utilized the Morris water maze spatial learning paradigm to systematically evaluate mild behavioral deficits that occur during the early stages of disease pathogenesis. Conventional behavioral analysis using this model indicates that spatial memory is intact at 2 months of age. In this study, we used an alternative method to analyze the behavior of mice, aiming to gain a better understanding of the nature of cognitive deficits by focusing on the unsuccessful trials during water maze learning rather than on the successful ones. APP/PS1 mice displayed a higher number of unsuccessful trials during the initial days of training, unlike their wild-type counterparts. However, with repeated trial and error, learning in APP/PS1 reached levels comparable to that of the wild-type mice during the later days of training. Individual APP/PS1 mice preferred a non-cognitive search strategy called circling, which led to abrupt learning transitions and an increased number of unsuccessful trials. These findings indicate the significance of subtle intermediate readouts as early indicators of conditions such as Alzheimer's disease.

5.
J Neurosci ; 28(47): 12500-9, 2008 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-19020042

RESUMO

Parkinson's disease (PD) is a progressive neurodegenerative disease characterized by the degeneration of the dopaminergic neurons in the substantia nigra pars compacta (SNpc). Activation of the mixed lineage kinase and c-Jun N-terminal kinase (JNK) has been reported in models of PD. Our focus was to discern whether distinct pathways were activated in cell-specific manner within the SNpc. We now demonstrate the selective phosphorylation of p38 MAP kinase within the dopaminergic neurons, whereas JNK activation occurs predominantly in the microglia. p38 activation results in downstream phosphorylation of p53 and increased p53 mediated transcription of Bax and Puma in the ventral midbrain. Treatment with p38 inhibitor, SB239063 protected primary dopaminergic neurons derived from human progenitor cells from MPP(+) mediated cell death and prevented the downstream phosphorylation of p53 and its translocation to the nucleus in vivo, in the ventral midbrain. The increased staining of phosphorylated p38 in the surviving neurons of SNpc in human brain sections from patients with PD and in MPTP treated mice but not in the ventral tegmental area provides further evidence suggesting a role for p38 in the degeneration of dopaminergic neurons of SNpc. We thus demonstrate the cell specific activation of MAP kinase pathways within the SNpc after MPTP treatment emphasizing the role of multiple signaling cascades in the pathogenesis and progression of the disease. Selective inhibitors of p38 may therefore, help preserve the surviving neurons in PD and slow down the disease progression.


Assuntos
Dopamina/metabolismo , Intoxicação por MPTP/patologia , Neurônios/metabolismo , Substância Negra/patologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/administração & dosagem , Análise de Variância , Animais , Benzotiazóis/farmacologia , Encéfalo , Células Cultivadas , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feto/citologia , Humanos , Imidazóis , MAP Quinase Quinase 4/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doença de Parkinson/patologia , Transporte Proteico/efeitos dos fármacos , Pirimidinas , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Substância Negra/metabolismo , Fatores de Tempo , Tolueno/análogos & derivados , Tolueno/farmacologia , Tirosina 3-Mono-Oxigenase/metabolismo , Regulação para Cima/efeitos dos fármacos
6.
Antioxid Redox Signal ; 31(18): 1321-1338, 2019 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-31617375

RESUMO

Aims: Reactive oxygen species (ROS) generated during Alzheimer's disease (AD) pathogenesis through multiple sources are implicated in synaptic pathology observed in the disease. We have previously shown F-actin disassembly in dendritic spines in early AD (34). The actin cytoskeleton can be oxidatively modified resulting in altered F-actin dynamics. Therefore, we investigated whether disruption of redox signaling could contribute to actin network disassembly and downstream effects in the amyloid precursor protein/presenilin-1 double transgenic (APP/PS1) mouse model of AD. Results: Synaptosomal preparations from 1-month-old APP/PS1 mice showed an increase in ROS levels, coupled with a decrease in the reduced form of F-actin and increase in glutathionylated synaptosomal actin. Furthermore, synaptic glutaredoxin 1 (Grx1) and thioredoxin levels were found to be lowered. Overexpressing Grx1 in the brains of these mice not only reversed F-actin loss seen in APP/PS1 mice but also restored memory recall after contextual fear conditioning. F-actin levels and F-actin nanoarchitecture in spines were also stabilized by Grx1 overexpression in APP/PS1 primary cortical neurons, indicating that glutathionylation of F-actin is a critical event in early pathogenesis of AD, which leads to spine loss. Innovation: Loss of thiol/disulfide oxidoreductases in the synapse along with increase in ROS can render F-actin nanoarchitecture susceptible to oxidative modifications in AD. Conclusions: Our findings provide novel evidence that altered redox signaling in the form of S-glutathionylation and reduced Grx1 levels can lead to synaptic dysfunction during AD pathogenesis by directly disrupting the F-actin nanoarchitecture in spines. Increasing Grx1 levels is a potential target for novel disease-modifying therapies for AD.


Assuntos
Actinas/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Disfunção Cognitiva/metabolismo , Modelos Animais de Doenças , Glutarredoxinas/metabolismo , Animais , Células Cultivadas , Glutarredoxinas/análise , Glutarredoxinas/genética , Masculino , Camundongos , Camundongos Transgênicos , Oxirredução , Presenilina-1/metabolismo , Espécies Reativas de Oxigênio/análise , Espécies Reativas de Oxigênio/metabolismo
7.
FASEB J ; 21(9): 2226-36, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17369508

RESUMO

Parkinson's disease (PD), a neurodegenerative disorder, causes severe motor impairment due to loss of dopaminergic neurons in substantia nigra pars compacta (SNpc). MPTP, a neurotoxin that causes dopaminergic cell loss in mice, was used in an animal model to study the pathogenic mechanisms leading to neurodegeneration. We observed the activation of apoptosis signal regulating kinase (ASK1, MAPKKK) and phosphorylation of its downstream targets MKK4 and JNK, 12 h after administration of a single dose of MPTP. Further, Daxx, the death-associated protein, translocated to the cytosol selectively in SNpc neurons seemingly due to MPTP mediated down-regulation of DJ-1, the redox-sensitive protein that binds Daxx in the nucleus. Coadministration of alpha-lipoic acid (ALA), a thiol antioxidant, abolished the activation of ASK1 and phosphorylation of downstream kinases, MKK4, and JNK and prevented the down-regulation of DJ-1 and translocation of Daxx to the cytosol seen after MPTP. ALA also attenuated dopaminergic cell loss in SNpc seen after subchronic MPTP treatment. Our studies demonstrate for the first time that MPTP triggers death signaling pathway by activating ASK1 and translocating Daxx, in vivo, in dopaminergic neurons in SNpc of mice and thiol antioxidants, such as ALA terminate this cascade and afford neuroprotection.


Assuntos
Antioxidantes/uso terapêutico , Antiparkinsonianos/uso terapêutico , Proteínas de Transporte/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , MAP Quinase Quinase Quinase 5/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Intoxicação por MPTP/tratamento farmacológico , Proteínas do Tecido Nervoso/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Proteínas Nucleares/metabolismo , Transtornos Parkinsonianos/tratamento farmacológico , Substância Negra/efeitos dos fármacos , Ácido Tióctico/uso terapêutico , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacocinética , Acetilcisteína/farmacologia , Alcinos/farmacologia , Animais , Antioxidantes/farmacologia , Antiparkinsonianos/farmacologia , Biotransformação , Núcleo Celular/metabolismo , Proteínas Correpressoras , Cistationina gama-Liase/antagonistas & inibidores , Citosol/metabolismo , Dopamina/análise , Avaliação Pré-Clínica de Medicamentos , Complexo I de Transporte de Elétrons/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Glutationa/análise , Glicina/análogos & derivados , Glicina/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase 4/metabolismo , Intoxicação por MPTP/metabolismo , Masculino , Mesencéfalo/química , Camundongos , Camundongos Endogâmicos C57BL , Chaperonas Moleculares , Neurônios/química , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Proteínas Oncogênicas/biossíntese , Proteínas Oncogênicas/genética , Transtornos Parkinsonianos/metabolismo , Peroxirredoxinas , Fosforilação , Proteína Desglicase DJ-1 , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Substância Negra/metabolismo , Ácido Tióctico/farmacologia
8.
Nat Neurosci ; 21(9): 1290, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30013169

RESUMO

In the version of this article initially published, the right panel in Fig. 2b was duplicated from the corresponding panel in Fig. 2c, and some data points in Fig. 3b were duplicated from Fig. 3a. None of the conclusions in the paper are affected. The errors have been corrected in the HTML and PDF versions of the article, and source data have been posted for the revised panels. The original and corrected figures are shown in the accompanying Author Correction.

9.
Brain Res ; 1185: 8-17, 2007 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-17961515

RESUMO

Oxidative stress and mitochondrial dysfunction caused by loss of complex I activity are presumed to be primary events leading to neurodegeneration in Parkinson's disease. Mitochondrial glutaredoxin (Grx2), a glutathione-dependent thiol disulfide oxidoreductase helps maintain redox homeostasis in the mitochondria. We therefore, examined the constitutive expression of Grx2 in brain and its role in MPTP-mediated mitochondrial dysfunction in the extrapyramidal system. Grx2 is constitutively expressed in both neuron and glia in mouse and human brain including the neurons in human substantia nigra. Grx2 mRNA and protein were transiently upregulated in midbrain and striatum 1 h but not 4 h after a single dose of MPTP. Downregulation of Grx2 using antisense oligonucleotides, in vivo, in mouse brain resulted in partial loss of complex I activity indicating that Grx2 may help maintain complex I function in the mitochondria. Further, overexpression of Grx2 abolished MPP(+)-mediated toxicity in vitro in neuroblastoma cells. Our results demonstrate the probable role of Grx2 in maintenance of the redox milieu in mitochondria and its potential neuroprotective role in preserving mitochondrial integrity in neurodegenerative diseases, such as Parkinson's disease.


Assuntos
Encéfalo/metabolismo , Glutarredoxinas/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Adulto , Idoso , Animais , Encéfalo/efeitos dos fármacos , Linhagem Celular Transformada , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Marcação In Situ das Extremidades Cortadas/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NADH NADPH Oxirredutases/metabolismo , Neurotoxinas/farmacologia , Oligonucleotídeos Antissenso/farmacologia , Mudanças Depois da Morte , Fatores de Tempo , Transfecção/métodos , Ubiquinona/metabolismo
10.
Nat Neurosci ; 19(3): 454-64, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26807952

RESUMO

Long-term consolidation of memories depends on processes occurring many hours after acquisition. Whether this involves plasticity that is specifically required for long-term consolidation remains unclear. We found that learning-induced plasticity of local parvalbumin (PV) basket cells was specifically required for long-term, but not short/intermediate-term, memory consolidation in mice. PV plasticity, which involves changes in PV and GAD67 expression and connectivity onto PV neurons, was regulated by cAMP signaling in PV neurons. Following induction, PV plasticity depended on local D1/5 dopamine receptor signaling at 0-5 h to regulate its magnitude, and at 12-14 h for its continuance, ensuring memory consolidation. D1/5 dopamine receptor activation selectively induced DARPP-32 and ERK phosphorylation in PV neurons. At 12-14 h, PV plasticity was required for enhanced sharp-wave ripple densities and c-Fos expression in pyramidal neurons. Our results reveal general network mechanisms of long-term memory consolidation that requires plasticity of PV basket cells induced after acquisition and sustained subsequently through D1/5 receptor signaling.


Assuntos
Consolidação da Memória , Memória de Longo Prazo/fisiologia , Plasticidade Neuronal/fisiologia , Parvalbuminas/metabolismo , Receptores de Dopamina D1/fisiologia , Receptores de Dopamina D5/fisiologia , Transdução de Sinais , Animais , AMP Cíclico/fisiologia , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glutamato Descarboxilase/metabolismo , Memória de Curto Prazo/fisiologia , Camundongos , Neurônios/metabolismo , Neurônios/fisiologia , Fatores de Tempo
11.
Free Radic Biol Med ; 87: 312-25, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26164633

RESUMO

Activation of apoptosis signal-regulating kinase 1 (ASK1)-p38 MAPK death signaling cascade is implicated in the death of dopaminergic neurons in substantia nigra in Parkinson's disease (PD). We investigated upstream activators of ASK1 using an MPTP mouse model of parkinsonism and assessed the temporal cascade of death signaling in ventral midbrain (VMB) and striatum (ST). MPTP selectively activated ASK1 and downstream p38 MAPK in a time-dependent manner in VMB alone. This occurred through selective protein thiol oxidation of the redox-sensitive thiol disulfide oxidoreductase, thioredoxin (Trx1), resulting in release of its inhibitory association with ASK1, while glutathione-S-transferase µ 1 (GSTM1) remained in reduced form in association with ASK1. Levels of tumor necrosis factor (TNF), a known activator of ASK1, increased early after MPTP in VMB. Protein covariation network analysis (PCNA) using protein states as nodes revealed TNF to be an important node regulating the ASK1 signaling cascade. In confirmation, blocking MPTP-mediated TNF signaling through intrathecal administration of TNF-neutralizing antibody prevented Trx1 oxidation and downstream ASK1-p38 MAPK activation. Averting an early increase in TNF, which leads to protein thiol oxidation resulting in activation of ASK1-p38 signaling, may be critical for neuroprotection in PD. Importantly, network analysis can help in understanding the cause/effect relationship within protein networks in complex disease states.


Assuntos
MAP Quinase Quinase Quinase 5/metabolismo , Doença de Parkinson/tratamento farmacológico , Tiorredoxinas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/administração & dosagem , Animais , Anticorpos Neutralizantes/administração & dosagem , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Modelos Animais de Doenças , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Mesencéfalo/patologia , Camundongos , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo , Substância Negra/patologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/imunologia
13.
Neurotox Res ; 16(2): 116-26, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19526288

RESUMO

Incidence of Parkinson's disease (PD) is lower in women compared to men (1:1.46), which is reflected in animal models. However, precise mechanisms are unclear. Administration of MPTP (1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine) to female mice does not lead to mitochondrial complex I inhibition as seen in males and the progressive dopaminergic cell loss in substantia nigra (SNpc) is significantly attenuated. Redox driven apoptotic signaling pathways regulated by thiol disulfide oxidoreductase(s) have been implicated in the neurodegeneration seen in PD. Oxidation of thioredoxin leads to activation of apoptosis signal regulating kinase 1 (ASK1; MAPKKK) initiating cell death cascade through MAP kinase(s). Higher constitutive expression of enzymes involved in cellular redox maintenance, such as glutathione reductase, thioredoxin, and thioredoxin reductase is observed in female brain. Exposure to MPTP activates ASK1 in male but not in female mice. Higher expression of Trx in females potentially prevents ASK1 activation. Downstream of ASK1, phosphorylation of p38 MAP kinase is seen in male but not female mice. Expression of DJ-1, the redox sensing protein is higher in females and the loss of nuclear DJ-1, followed by translocation of Daxx (death associated protein) from the nucleus to the cytosol, which promotes ASK1 mediated death cascade is not seen in females. The enzymes involved in redox maintenance potentially could play a crucial role in preventing the activation of redox driven death signaling cascade and offer neuroprotection. Theraupeutic strategies that help maintain redox homeostasis may help prevent the progressive neurodegeneration seen in PD.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , MAP Quinase Quinase Quinase 5/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neurotoxinas/farmacologia , Oxirredução , Animais , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Proteínas Correpressoras , Dopamina/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glutationa Redutase/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Chaperonas Moleculares , NAD/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Oxirredução/efeitos dos fármacos , Peroxirredoxinas , Proteína Desglicase DJ-1 , Transporte Proteico/efeitos dos fármacos , Fatores Sexuais , Tiorredoxina Dissulfeto Redutase/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA