Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neurochem Res ; 49(6): 1450-1467, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38720090

RESUMO

Wernicke's encephalopathy (WE) is a major central nervous system disorder resulting from thiamine deficiency (TD) in which a number of brain regions can develop serious damage including the thalamus and inferior colliculus. Despite decades of research into the pathophysiology of TD and potential therapeutic interventions, little progress has been made regarding effective treatment following the development of brain lesions and its associated cognitive issues. Recent developments in our understanding of stem cells suggest they are capable of repairing damage and improving function in different maladys. This article puts forward the case for the potential use of stem cell treatment as a therapeutic strategy in WE by first examining the effects of TD on brain functional integrity and its consequences. The second half of the paper will address the future benefits of treating TD with these cells by focusing on their nature and their potential to effectively treat neurodegenerative diseases that share some overlapping pathophysiological features with TD. At the same time, some of the obstacles these cells will have to overcome in order to become a viable therapeutic strategy for treating this potentially life-threatening illness in humans will be highlighted.


Assuntos
Transplante de Células-Tronco , Deficiência de Tiamina , Encefalopatia de Wernicke , Humanos , Deficiência de Tiamina/terapia , Deficiência de Tiamina/complicações , Animais , Encefalopatia de Wernicke/etiologia , Encefalopatia de Wernicke/terapia , Transplante de Células-Tronco/métodos , Encéfalo/patologia , Encéfalo/metabolismo
2.
Metab Brain Dis ; 36(2): 239-246, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33245475

RESUMO

Thiamine deficiency (TD) results in focal lesions in several regions of the rat brain including the thalamus and inferior colliculus. Since alterations in blood-brain barrier (BBB) integrity may play a role in this damage, we have examined the influence of TD on the unidirectional blood-to-brain transfer constant (Ki) of the low molecular weight species α-aminoisobutyric acid (AIB) in vulnerable and non-vulnerable brain regions at different stages during progression of the disorder, and following its reversal with thiamine. Analysis of the regional distribution of Ki values showed early (day 10) increased transfer of [14C]-AIB across the BBB in the vulnerable medial thalamus as well as the non-vulnerable caudate and hippocampus. At the acute symptomatic stage (day 14), more widespread BBB permeability changes were detected in most areas including the lateral thalamus, inferior colliculus, and non-vulnerable cerebellum and pons. Twenty-four hours following thiamine replenishment, a heterogeneous pattern of increased BBB permeability was observed in which many structures maintained increased uptake of [14C]-AIB. No increase in the [3H]-dextran space, a marker of intravascular volume, was detected in brain regions during the progress of TD, suggesting that BBB permeability to this large tracer was unaffected. These results indicate that BBB opening i) occurs early during TD, ii) is not restricted to vulnerable areas of the brain, iii) is progressive, iv) persists for at least 24 h following treatment with thiamine, and v) is likely selective in nature, depending on the molecular species being transported.


Assuntos
Ácidos Aminoisobutíricos/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Deficiência de Tiamina/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Masculino , Permeabilidade/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
3.
Neurochem Int ; 112: 134-145, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29032009

RESUMO

Development of manganism (also known as manganese neurotoxicity) is a major complication of manganese exposure in which neurological dysfunction is linked to accumulation of the metal in brain. Due to neuronal cell death in basal ganglia structures, particularly the globus pallidus, functional recovery is limited. Bearing a resemblance to Parkinson's disease, effective treatment for manganism is currently limited. However, the rapidly developing field of stem cell research offers new hope for the treatment of illnesses in which neurodegeneration is a major feature. The first part of this review will focus on the clinical features and pathophysiology of cerebral damage resulting from exposure to manganese, including the role of astrocytes, disruption of energy metabolism, involvement of oxidative stress, excitotoxicity, and inflammation, with the second part exploring how stem cells may provide an important therapeutic strategy for patients with this major neurologic disorder.


Assuntos
Intoxicação por Manganês/terapia , Estresse Oxidativo/fisiologia , Transplante de Células-Tronco/métodos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Humanos , Intoxicação por Manganês/metabolismo , Transplante de Células-Tronco/tendências , Resultado do Tratamento
4.
Neurosci Lett ; 656: 58-64, 2017 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-28648459

RESUMO

Hepatic encephalopathy (HE) represents a serious complication of acute liver failure (ALF) in which cerebral edema leading to brainstem herniation as a result of increased intracranial hypertension is a major consequence. Long non-coding RNAs (lncRNAs) play a significant role in coordinating gene expression, with recent studies indicating an influence in the pathogenesis of several diseases. To investigate their involvement in the cerebral pathophysiology of ALF, we profiled the expression of lncRNAs in the frontal cortex of mice at coma stage following treatment with the hepatotoxin azoxymethane. Of the 35,923 lncRNAs profiled using microarrays, 868 transcripts were found to be differentially expressed in the ALF-treated group compared to the sham control group. Of these, 382 lncRNAs were upregulated and 486 lncRNAs downregulated. Pathway analysis revealed these lncRNAs target a number of biological and molecular pathways that include cytokine-cytokine receptor interaction, the mitogen activated protein kinase signaling pathway, the insulin signaling pathway, and the nuclear factor-κB signaling pathway. False discovery rate adjustment identified 9 upregulated lncRNAs, 2 of which are associated with neuroepithelial transforming gene 1 (NET1) and the monocarboxylate transporter 2 (Slc16a7), potential contributors to astrocyte cytoskeletal disruption/swelling and lactate production, respectively. Our findings suggest an important role for lncRNAs in the brain in ALF in relation to inflammation, neuropathology, and in terms of the functional basis of HE. Further work on these non-coding RNAs may lead to new therapeutic approaches for the treatment and management of cerebral dysfunction resulting from this potentially life-threatening disorder.


Assuntos
Córtex Cerebral/metabolismo , Falência Hepática Aguda/genética , RNA Longo não Codificante/metabolismo , Animais , Falência Hepática Aguda/metabolismo , Masculino , Camundongos Endogâmicos C57BL
5.
Neurosci Lett ; 642: 163-167, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28109775

RESUMO

Recent clinical trials suggest that patients with myelofibrosis can develop Wernicke's encephalopathy (WE) when treated with fedratinib, a specific Janus kinase-2 (JAK-2) inhibitor. To investigate this issue, we have examined (1) if fedratinib can produce or alter the course of this disorder, (2) its effects on thiamine-dependent enzyme activity and thiamine status, and (3) its influence on the uptake of thiamine. Animals administered fedratinib for 28days at a comparable dose used to treat human cases of myelofibrosis showed no evidence of clinical signs of thiamine deficiency (TD). Rats treated with a combination of fedratinib and TD exhibited no neurological differences in their progress to the symptomatic stage when compared to thiamine-deficient animals only. Treatment with the JAK-2 inhibitor did not compromise erythrocyte transketolase activity, and thiamine status was not affected in a major way unlike animals with TD. In addition, treatment of cultured astrocytes with fedratinib did not diminish the uptake of thiamine into these cells. Our findings suggest that treatment with fedratinib does not lead to or alter the progress of TD, and do not support the notion that administration of this JAK-2 inhibitor directly results in the development of WE due to inhibition of thiamine transport. Known adverse effects of fedratinib involving compromised gastrointestinal function may be an important indirect contributing factor to previously reported cases of WE in patients with myelofibrosis.


Assuntos
Janus Quinase 2/antagonistas & inibidores , Pirrolidinas , Sulfonamidas , Deficiência de Tiamina/induzido quimicamente , Encefalopatia de Wernicke/induzido quimicamente , Animais , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley , Tiamina/sangue , Deficiência de Tiamina/sangue
6.
Front Neurol ; 7: 118, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27602016

RESUMO

The vegetative state (VS), also known as "unresponsive wakefulness syndrome," is considered one of the most devastating outcomes of acquired brain injury. While diagnosis of this condition is generally well-defined clinically, patients often appear to be awake despite an absence of behavioral signs of awareness, which to the family can be confusing, leading them to believe the loved one is aware of their surroundings. This inequality of agreement can be very distressing. Currently, no cure for the VS is available; as a result, patients may remain in this condition for the rest of their lives, which in some cases amount to decades. Recent advances in stem cell approaches for the treatment of other neurological conditions may now provide an opportunity to intervene in this syndrome. This mini review will address the development of VS, its diagnosis, affected cerebral structures, and the underlying basis of how stem cells can offer therapeutic promise that would take advantage of the often long-term features associated with this maladie to effect a repair of the severely damaged circuitry. In addition, current limitations of this treatment strategy, including a lack of animal models, few long-term clinical studies that might identify benefits of stem cell treatment, and the potential for development of tumors are considered.

7.
Neurochem Res ; 40(2): 353-61, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25297573

RESUMO

Thiamine is an essential vitamin that is necessary to maintain the functional integrity of cells in the brain. Its deficiency is the underlying cause of Wernicke's encephalopathy (WE), a disorder primarily associated with, but not limited to, chronic alcoholism. Thiamine deficiency leads to the development of impaired energy metabolism due to mitochondrial dysfunction in focal regions of the brain resulting in cerebral vulnerability. The consequences of this include oxidative stress, excitotoxicity, inflammatory responses, decreased neurogenesis, blood-brain barrier disruption, lactic acidosis and a reduction in astrocyte functional integrity involving a loss of glutamate transporters and other astrocyte-specific proteins which together contribute in a major way to the resulting neurodegeneration. Exactly how these factors acting in concert lead to the demise of neurons is unclear. In this review we reassess their relative importance in the light of more recent findings and discuss therapeutic possibilities that may provide hope for the future for individuals with WE.


Assuntos
Deficiência de Tiamina/fisiopatologia , Deficiência de Tiamina/terapia , Animais , Glutamatos/fisiologia , Humanos , Estresse Oxidativo
10.
Metab Brain Dis ; 29(4): 1061-8, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24929329

RESUMO

Thiamine deficiency (TD) is the underlying cause of Wernicke's encephalopathy (WE), an acute neurological disorder characterized by structural damage to key periventricular structures in the brain. Increasing evidence suggests these focal histological lesions may be representative of a gliopathy in which astrocyte-related changes are a major feature of the disorder. These changes include a loss of the glutamate transporters GLT-1 and GLAST concomitant with elevated interstitial glutamate levels, lowered brain pH associated with increased lactate production, decreased levels of GFAP, reduction in the levels of glutamine synthetase, swelling, alterations in levels of aquaporin-4, and disruption of the blood-brain barrier. This review focusses on how these manifestations contribute to the pathophysiology of TD and possibly WE.


Assuntos
Astrócitos/fisiologia , Deficiência de Tiamina/fisiopatologia , Sistema X-AG de Transporte de Aminoácidos/fisiologia , Animais , Transporte Biológico , Barreira Hematoencefálica , Encéfalo/patologia , Modelos Animais de Doenças , Transportador 2 de Aminoácido Excitatório/fisiologia , Ácido Glutâmico/metabolismo , Humanos , Complexo Cetoglutarato Desidrogenase/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo , Piritiamina/toxicidade , Deficiência de Tiamina/induzido quimicamente , Deficiência de Tiamina/metabolismo , Encefalopatia de Wernicke/etiologia , Encefalopatia de Wernicke/metabolismo , Encefalopatia de Wernicke/fisiopatologia
11.
Metab Brain Dis ; 29(4): 891-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24861182

RESUMO

The mechanisms that promote brain dysfunction after acute liver failure (ALF) are not clearly understood. The small noncoding RNAs known as microRNAs (miRNAs) significantly control mRNA translation and thus normal and pathological functions in the mammalian body. To understand their significance in ALF, we currently profiled the expression of miRNAs in the cerebral cortex of mice sacrificed at coma stage following treatment with azoxymethane. Of the 470 miRNAs profiled using microarrays, 37 were significantly altered (20 up-and 17 down-regulated) in their expression in the ALF group compared to sham group. In silico analysis showed that the ALF-responsive miRNAs target on average 231 mRNAs/miRNA (range: 3 to 840 targets). Pathways analysis showed that many miRNAs altered after ALF target multiple mRNAs that are part of various biological and molecular pathways. Glutamatergic synapse, Wnt signaling, MAP-kinase signaling, axon guidance, PI3-kinase-AKT signaling, T-cell receptor signaling and ubiquitin-mediated proteolysis are the top pathways targeted by the ALF-sensitive miRNAs. At least 28 ALF-responsive miRNAs target each of the above pathways. We hypothesize that alterations in miRNAs and their down-stream mRNAs of signaling pathways might play a role in the induction and progression of neurological dysfunction observed during ALF.


Assuntos
Córtex Cerebral/metabolismo , Perfilação da Expressão Gênica , Encefalopatia Hepática/metabolismo , Falência Hepática/complicações , MicroRNAs/biossíntese , Animais , Azoximetano/toxicidade , Encefalopatia Hepática/etiologia , Falência Hepática/induzido quimicamente , Falência Hepática/metabolismo , Masculino , Redes e Vias Metabólicas/genética , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , RNA Mensageiro/genética , Transdução de Sinais/genética
12.
Biochem Biophys Res Commun ; 448(3): 335-41, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24735535

RESUMO

Loss of astrocytic glutamate transporters is a major feature of both thiamine deficiency (TD) and Wernicke's encephalopathy. However, the underlying basis of this process is not well understood. In the present study we have investigated the possibility of release of astrocytic soluble factors that might be involved in the regulation of the glutamate transporter GLT-1b in these cells. Treatment of naïve astrocytes with conditioned media from astrocytes exposed to TD conditions resulted in a progressive decrease in glutamate uptake over 24 h. Immunoblotting and flow cytometry measurements indicated this was accompanied by a 20-40% loss of GLT-1b. Astrocytes exposed to either TD or TD conditioned media showed increased disruption of mitochondrial membrane potential compared to control cells, and treatment of astrocytes with TD resulted in an increase in the pro-inflammatory cytokine TNF-α and elevated levels of phospho-IκB fragment, indicative of increased activation of NF-κB. Inhibition of TNF-α activity with the use of a neutralizing antibody blocked the increased NF-κB activation, while inhibition of NF-κB ameliorated the decrease in GLT-1b and reversed the decrease in glutamate uptake occurring with TD treatment. Together, these findings indicate that astrocytes exposed to TD conditions show responses suggesting that soluble factors released by these cells under conditions of TD play a regulatory role in terms of glutamate transport function and mitochondrial integrity.


Assuntos
Astrócitos/metabolismo , Transportador 2 de Aminoácido Excitatório/genética , Transportador 2 de Aminoácido Excitatório/metabolismo , Deficiência de Tiamina/metabolismo , Animais , Células Cultivadas , Meios de Cultivo Condicionados , Citocinas/metabolismo , Regulação para Baixo , Ácido Glutâmico/metabolismo , Mediadores da Inflamação/metabolismo , Potencial da Membrana Mitocondrial , NF-kappa B/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratos , Deficiência de Tiamina/complicações , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo , Encefalopatia de Wernicke/etiologia , Encefalopatia de Wernicke/metabolismo
13.
Metab Brain Dis ; 29(1): 145-52, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24078061

RESUMO

Thiamine deficiency (TD) leads to Wernicke's encephalopathy (WE), in which focal histological lesions occur in periventricular areas of the brain. Recently, impaired neurogenesis has been reported in the hippocampus during the dietary form of TD, and in pyrithiamine-induced TD (PTD), a well-characterized model of WE. To further characterize the consequences of PTD on neural stem/progenitor cell (NSPC) activity, we have examined the effect of this treatment in the rat on both the subventricular zone (SVZ) of the rostral lateral ventricle and subgranular layer (SGL) of the hippocampus, and in the thalamus and inferior colliculus, two vulnerable brain regions in this disorder. In both the SVZ and SGL, PTD led to a decrease in the numbers of bromodeoxyuridine-stained cells, indicating that proliferation of NSPCs destined for neurogenesis in these areas was reduced. Doublecortin (DCX) immunostaining in the SGL was decreased, indicating a reduction in neuroblast formation, consistent with impaired NSPC activity. DCX labeling was not apparent in focal areas of vulnerability. In the thalamus, proliferation of cells was absent while in the inferior colliculus, numerous actively dividing cells were apparent, indicative of a differential response between these two brain regions. Exposure of cultured neurospheres to PTD resulted in decreased proliferation of NSPCs, consistent with our in vivo findings. Together, these results indicate that PTD considerably affects cell proliferation and neurogenesis activity in both neurogenic areas and parts of the brain known to display structural and functional vulnerability, confirming and extending recent findings on the effects of TD on neurogenesis. Future use of NSPCs in vitro may allow a closer and more detailed examination of the mechanism(s) underlying inhibition of these cells during TD.


Assuntos
Encéfalo/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Piritiamina/toxicidade , Encefalopatia de Wernicke/patologia , Animais , Encéfalo/patologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Replicação do DNA/efeitos dos fármacos , Modelos Animais de Doenças , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Colículos Inferiores/efeitos dos fármacos , Colículos Inferiores/patologia , Ventrículos Laterais/efeitos dos fármacos , Ventrículos Laterais/patologia , Masculino , Proteínas Associadas aos Microtúbulos/análise , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/patologia , Neuropeptídeos/análise , Ratos , Ratos Sprague-Dawley , Tálamo/efeitos dos fármacos , Tálamo/patologia , Encefalopatia de Wernicke/induzido quimicamente
14.
Neurochem Int ; 62(5): 796-802, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23333339

RESUMO

Thiamine (vitamin B1) deficiency, the underlying cause of Wernicke-Korsakoff syndrome, is associated with the development of focal neuronal loss in vulnerable areas of the brain. Although the actual mechanism(s) that lead to the selective histological lesions characteristic of this disorder remain unresolved, oxidative stress has been shown to play a major role in its pathophysiology. In this review, the multifactorial influence of oxidative stress on a variety of processes known to take part in the development of structural lesions in TD including excitotoxicity, neuroinflammation, blood-brain barrier integrity, mitochondrial integrity, apoptosis, nucleic acid function, and neural stem cells will be discussed, and therapeutic strategies undertaken for treating neurodegeneration examined which may have an impact on the future treatment of this important vitamin deficiency.


Assuntos
Estresse Oxidativo , Deficiência de Tiamina/metabolismo , Animais , Apoptose , Barreira Hematoencefálica , Humanos , Mitocôndrias/metabolismo
15.
Neurochem Int ; 58(5): 558-63, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21256174

RESUMO

Downregulation of astrocytic glutamate transporters is a feature of thiamine deficiency (TD), the underlying cause of Wernicke's encephalopathy, and plays a major role in its pathophysiology. Recent investigations suggest that ceftriaxone, a ß-lactam antibiotic, stimulates GLT-1 expression and confers neuroprotection against ischemic and motor neuron degeneration. Thus, ceftriaxone treatment may be a protective strategy against excitotoxic conditions. In the present study, we examined the effects of ceftriaxone on the glutamate transporter splice-variant GLT-1b in rats with TD and in cultured astrocytes under TD conditions. Our results indicate that ceftriaxone protects against loss of GLT-1b levels in the inferior colliculus during TD, but with no significant effect in the thalamus and frontal cortex by immunoblotting and immunohistochemistry. Ceftriaxone also normalized the loss of GLT-1b in astrocyte cultures under conditions of TD. These results suggest that ceftriaxone has the ability to increase GLT-1b levels in astrocytes during TD, and may be an important pharmacological strategy for the treatment of excitotoxicity in this disorder.


Assuntos
Ceftriaxona/uso terapêutico , Transportador 2 de Aminoácido Excitatório/deficiência , Colículos Inferiores/metabolismo , Deficiência de Tiamina/metabolismo , Deficiência de Tiamina/prevenção & controle , Animais , Ceftriaxona/farmacologia , Células Cultivadas , Transportador 2 de Aminoácido Excitatório/genética , Colículos Inferiores/efeitos dos fármacos , Masculino , Isoformas de Proteínas/deficiência , Isoformas de Proteínas/genética , Ratos , Ratos Sprague-Dawley , Deficiência de Tiamina/genética
16.
Biochem Biophys Res Commun ; 404(1): 228-32, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21110949

RESUMO

Complexins are a family of SNARE complex-binding proteins which regulate neurotransmitter release by playing a crucial role in triggering fast exocytosis at the synapse. Current evidence indicates astrocytes can release glutamate via a vesicular mechanism similar to that at nerve terminals and thereby modulate synaptic activity. In addition, components of the biochemical machinery associated with synaptic release have been identified in these cells. However, whether complexins are also present in astrocytes and may therefore participate in the vesicular release of glutamate is a key issue that is yet to be determined. In the present study we therefore examined if astrocytes express complexin I (Cpx I) and/or complexin II (Cpx II). Our results indicate these cells contain Cpx II but not Cpx I in primary culture. In addition, serum deprivation for 24 h led to a 2.6-fold increase in Cpx II, suggesting this protein is responsive to insults. These findings point to Cpx II being a likely key modulator of synaptic activity at the level of these glial cells. Given the considered involvement of complexins in neurologic and psychiatric illness, astrocytic Cpx II represents a potentially important therapeutic target for the future treatment of such maladies.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Astrócitos/metabolismo , Ácido Glutâmico/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Células Cultivadas , Ratos , Sinapses/metabolismo
17.
Neurochem Int ; 58(3): 248-60, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21130821

RESUMO

Emerging evidence suggests that thiamine deficiency (TD), the cause of Wernicke's encephalopathy, produces alterations in brain function and structural damage that closely model a number of maladies in which neurodegeneration is a characteristic feature, including Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease, multiple sclerosis, along with alcoholic brain disease, stroke, and traumatic brain injury. Impaired oxidative metabolism in TD due to decreased activity of thiamine-dependent enzymes leads to a multifactorial cascade of events in the brain that include focal decreases in energy status, oxidative stress, lactic acidosis, blood-brain barrier disruption, astrocyte dysfunction, glutamate-mediated excitotoxicity, amyloid deposition, decreased glucose utilization, immediate-early gene induction, and inflammation. This review describes our current understanding of the basis of these abnormal processes in TD, their interrelationships, and why this disorder can be useful for our understanding of how decreased cerebral energy metabolism can give rise to cell death in different neurodegenerative disease states.


Assuntos
Metabolismo Energético/fisiologia , Doenças Neurodegenerativas/metabolismo , Deficiência de Tiamina/metabolismo , Tiamina/metabolismo , Animais , Humanos , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/fisiopatologia , Oxirredução , Fosforilação Oxidativa/efeitos dos fármacos , Deficiência de Tiamina/complicações , Deficiência de Tiamina/fisiopatologia
18.
Biochem Biophys Res Commun ; 402(1): 123-8, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20932820

RESUMO

In Wernicke's encephalopathy and thiamine deficiency (TD), the cause of this brain disorder, development of inflammation is an important aspect of the disease process. How this pathological mechanism relates to the neurologic impairment associated with TD, however, remains unclear. A key feature of the inflammatory process is the activation of microglia. In the present study, we evaluated the role of microglial activation in the pathophysiology of TD by examining the relationship between levels of CD11b/c and CD68, two proteins associated with microglial activation, and neurological dysfunction under conditions of TD. Rats with TD showed large increases in expression of both CD11b/c and CD68 in the vulnerable thalamus and inferior colliculus, with no change in mRNA levels in the relatively non-vulnerable frontal cortex. These alterations in CD11b/c and CD68 expression were reflected in dramatic upregulation of both proteins by immunoblotting and immunohistochemical methods. Co-treatment of rats with TD and the anti-inflammatory drug minocycline prevented microglial activation, and onset of neurological changes, including loss of righting reflex, was delayed by approximately 39h, compared to animals with TD alone. In addition, co-treatment of rats with TD and N-acetylcysteine prevented the increase in CD11b/c and CD68, but did not alter the onset of neurological impairment. These results suggest that microglial activation plays a role in the development of neurological impairment in TD and possibly Wernicke's encephalopathy, and that while development of oxidative stress may be involved in microglial activation, the basis of this neurologic dysfunction is likely to be multifactorial in nature.


Assuntos
Encéfalo/fisiopatologia , Microglia/metabolismo , Deficiência de Tiamina/fisiopatologia , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/genética , Antígenos de Diferenciação Mielomonocítica/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Antígenos CD11/genética , Antígenos CD11/metabolismo , Masculino , Microglia/patologia , Ratos , Ratos Sprague-Dawley , Deficiência de Tiamina/metabolismo , Deficiência de Tiamina/patologia , Encefalopatia de Wernicke/metabolismo , Encefalopatia de Wernicke/patologia , Encefalopatia de Wernicke/fisiopatologia
19.
Neurochem Int ; 57(7): 830-7, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20816907

RESUMO

Wernicke's encephalopathy is a cerebral metabolic disorder caused by thiamine (vitamin B1) deficiency (TD). Neuropathologic consequences of TD include region-selective neuronal cell loss and blood-brain barrier (BBB) breakdown. Caveolin-1 is involved in the regulation of tight junction proteins and BBB permeability, and is modulated by oxidative stress, a feature of vulnerable brain regions in TD. We hypothesized that TD-related oxidative stress alters BBB integrity via induction of the caveolin-1 pathway. TD was induced in C57BL6 mice by treatment with a thiamine-deficient diet and administration of the thiamine antagonist pyrithiamine, in the absence or presence of the antioxidant N-acetylcysteine (NAC). A significant and focal increase in both caveolin-1 gene and protein expression was detected in the thalamus of thiamine-deficient mice, concomitant with IgG extravasation. Reduction of oxidative stress by NAC, as shown by normalization of reduced glutathione levels and attenuation of endothelial heme oxygenase-1 and nitric oxide synthase expression, resulted in prevention of the up-regulation of caveolin-1 in TD. Normalization of caveolin-1 levels by NAC was accompanied by a reduction in BBB breakdown, indicated by decreased IgG extravasation, normalization of occludin levels and prevention of matrix metalloproteinase-9 up-regulation. These findings demonstrate a role for caveolin-1 in TD pathogenesis, and suggest that oxidative stress contributes to BBB alterations in TD via modulation of this pathway.


Assuntos
Acetilcisteína/uso terapêutico , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Caveolina 1/biossíntese , Fármacos Neuroprotetores/metabolismo , Deficiência de Tiamina/metabolismo , Deficiência de Tiamina/prevenção & controle , Regulação para Cima/fisiologia , Acetilcisteína/farmacologia , Animais , Progressão da Doença , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/fisiologia , Distribuição Aleatória , Transdução de Sinais/fisiologia , Deficiência de Tiamina/patologia
20.
Glia ; 58(2): 148-56, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19565658

RESUMO

Wernicke encephalopathy (WE), a neurological disorder caused by thiamine deficiency (TD), is characterized by structural damage in brain regions that include the thalamus and cerebral cortex. The basis for these lesions is unclear, but may involve a disturbance of glutamatergic neurotransmission. We have therefore investigated levels of the astrocytic glutamate transporters EAAT1 and EAAT2 in order to evaluate their role in the pathophysiology of this disorder. Histological assessment of the frontal cortex revealed a significant loss of neurons in neuropathologically confirmed cases of WE compared with age-matched controls, concomitant with decreases in alpha-internexin and synaptophysin protein content of 67 and 52% by immunoblotting. EAAT2 levels were diminished by 71% in WE, with levels of EAAT1 also reduced by 62%. Loss of both transporter sites was confirmed by immunohistochemical methods. Development of TD in rats caused a profound loss of EAAT1 and EAAT2 in the thalamus accompanied by decreases in other astrocyte-specific proteins. Treatment of TD rats with N-acetylcysteine prevented the downregulation of EAAT2 in the medial thalamus, and ameliorated the loss of several other astrocyte proteins, concomitant with increased neuronal survival. Our results suggest that (1) loss of EAAT1 and EAAT2 glutamate transporters is associated with structural damage to the frontal cortex in patients with WE, (2) oxidative stress plays an important role in this process, and (3) TD has a profound effect on the functional integrity of astrocytes. Based on these findings, we recommend that early treatment using a combination of thiamine AND antioxidant approaches should be an important consideration in cases of WE.


Assuntos
Transportador 1 de Aminoácido Excitatório/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Proteínas de Transporte de Glutamato da Membrana Plasmática/metabolismo , Deficiência de Tiamina/fisiopatologia , Encefalopatia de Wernicke/fisiopatologia , Acetilcisteína/farmacologia , Adulto , Idoso , Animais , Astrócitos/metabolismo , Lobo Frontal/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley , Tálamo/metabolismo , Deficiência de Tiamina/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA