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











Base de dados
Intervalo de ano de publicação
1.
Behav Brain Res ; 416: 113568, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34499936

RESUMO

Hypobaric Hypoxia (HH) is known to cause oxidative stress in the brain that leads to spatial memory deficit and neurodegeneration. For decades therapeutic hypothermia is used to treat global and focal ischemia in preserving brain functions that proved to be beneficial in humans and rodents. Considering these previous reports, the present study was designed to establish the therapeutic potential of hypothermia preconditioning on HH induced spatial memory, biochemical and morphological changes in adult rats. Male Sprague Dawley rats were exposed to HH (7620 m, ~ 282 mmHg) for 1, 3 and 7 days with and without hypothermic preconditioning. Spatial learning memory was assessed by Morris water maze (MWM) test along with evaluation of hippocampal pyramidal neuron damage by histological study. Oxidative stress was measured by studying the levels of nitric oxide (NO), reactive oxygen species (ROS), lipid peroxidation (LPO), oxidized and reduced glutathione (GSSG and GSH). Results of MWM test indicated prolonged path length and latency to reach the platform in HH groups that regained to normal in cold pre-treated groups. A likely neurodegeneration was evident in HH groups that lessen in the cold pre-treated groups. Hypothermic preconditioning prevented spatial memory impairment and neurodegeneration in animals subjected to HH via decreasing the NO, ROS and LPO compared to control animals. The GSH level and GSH/GSSG ratio was found to be higher in preconditioned animals as compared to respective HH exposed animals, indicative of redox scavenging and restoration of hippocampal neuronal structure as well as spatial memory. Therefore, hypothermic preconditioning improves spatial memory deficit by reducing HH induced oxidative stress and hippocampal neurodegeneration, hence can be used as a multi-target prophylactic measure to combat HH induced neurodegeneration.


Assuntos
Hipocampo/fisiopatologia , Hipotermia/induzido quimicamente , Hipóxia Encefálica/fisiopatologia , Transtornos da Memória/fisiopatologia , Células Piramidais/patologia , Memória Espacial/fisiologia , Animais , Glutationa/metabolismo , Hipocampo/patologia , Hipóxia Encefálica/patologia , Peroxidação de Lipídeos/fisiologia , Masculino , Teste do Labirinto Aquático de Morris , Estresse Oxidativo/fisiologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio
2.
Biochim Biophys Acta Mol Basis Dis ; 1866(7): 165769, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32184133

RESUMO

Brain is well known for its disproportionate oxygen consumption and high energy-budget for optimal functioning. The decrease in oxygen supply to brain, thus, necessitates rapid activation of adaptive pathways - the absence of which manifest into vivid pathological conditions. Amongst these, oxygen sensing in glio-vascular milieu and H2S-dependent compensatory increase in cerebral blood flow (CBF) is a major adaptive response. We had recently demonstrated that the levels of H2S were significantly decreased during chronic hypobaric hypoxia (HH)-induced neuro-pathological effects. The mechanistic basis of this phenomenon, however, remained to be deciphered. We, here, describe experimental evidence for marked limitation of cysteine during HH - both in animal model as well as human volunteers ascending to high altitude. We show that the preservation of brain cysteine level, employing cysteine pro-drug (N-acetyl-L-cysteine, NAC), markedly curtailed effects of HH - not only on endogenous H2S levels but also, impairment of spatial reference memory in our animal model. We, further, present multiple lines of experimental evidence that the limitation of cysteine was causally governed by physiological propensity of brain to utilize cysteine, in cystathionine beta synthase (CBS)-dependent manner, past its endogenous replenishment potential. Notably, decrease in the levels of brain cysteine manifested despite positive effect (up-regulation) of HH on endogenous cysteine maintenance pathways and thus, qualifying cysteine as a conditionally essential nutrient (CEN) during HH. In brief, our data supports an adaptive, physiological role of CBS-mediated cysteine-utilization pathway - activated to increase endogenous levels of H2S - for optimal responses of brain to hypobaric hypoxia.


Assuntos
Doença da Altitude/metabolismo , Encéfalo/metabolismo , Cistationina beta-Sintase/genética , Cisteína/metabolismo , Sulfeto de Hidrogênio/metabolismo , Acetilcisteína/farmacologia , Adaptação Fisiológica , Adulto , Doença da Altitude/tratamento farmacológico , Doença da Altitude/genética , Doença da Altitude/patologia , Animais , Encéfalo/patologia , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/genética , Cistationina beta-Sintase/metabolismo , Modelos Animais de Doenças , Metabolismo Energético/genética , Humanos , Hipóxia/tratamento farmacológico , Hipóxia/genética , Hipóxia/metabolismo , Masculino , Consumo de Oxigênio/genética , Pró-Fármacos/farmacologia , Ratos , Adulto Jovem
3.
Brain Behav Immun ; 82: 129-144, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31408672

RESUMO

BACKGROUND: An association between neuroinflammation, reduced adult neurogenesis, and cognitive impairment has been established in sleep deprivation (SD). Complement receptors are expressed on neuronal and glial cells, thus, regulate the neuroinflammation, neurogenesis and learning/memory. However, understanding of the effect of SD on the brain-immune system interaction associated with cognitive dysfunction and its mechanisms is obscure. We hypothesized that complement activation induced changes in inflammatory and neurogenesis related proteins might be involved in the cognitive impairment during SD. METHODOLOGY: Adult male Sprague Dawley rats were used. Rats were sleep deprived for 48 h using a novel automated SD apparatus. Dosage of BrdU (50 mg/kg/day, i.p. in 0.07 N NaOH), complement C3a receptor antagonist (C3aRA; SB290157; 1 mg/kg/day, i.p.) in 1.16% v/v PBS and complement C5a receptor antagonist (C5aRA; W-54011; 1 mg/kg/day, i.p.) in normal saline were used. Rats were subjected to spatial memory evaluation following SD. Hippocampal tissue was collected for biochemical, molecular, and immunohistochemical studies. T-test and ANOVA were used for the statistical analysis. RESULTS: An up-regulation in the levels of complement components (C3, C5, C3a, C5a) and receptors (C3aR and C5aR) in hippocampus, displayed the complement activation during SD. Selective antagonism of C3aR/C5aR improved the spatial memory performance of sleep-deprived rats. C3aR antagonist (C3aRA) or C5aR antagonist (C5aRA) treatment inhibited the gliosis, maintained inflammatory cytokines balance in hippocampus during SD. Complement C3aR/C5aR antagonism improved hippocampal adult neurogenesis via up-regulating the BDNF level following SD. Administration of C3aRA and C5aRA significantly maintained synaptic homeostasis in hippocampus after SD. Gene expression analysis showed down-regulation in the mRNA levels of signal transduction pathways (Notch and Wnt), differentiation and axogenous proteins, which were found to be improved after C3aRA/C5aRA treatment. These findings were validated at protein and cellular level. Changes in the corticosterone level and ATP-adenosine-NO pathway were established as the key mechanisms underlying complement activation mediated consequences of SD. CONCLUSION: Our study suggests complement (C3a-C3aR and C5a-C5aR) activation as the novel mechanism underlying spatial memory impairment via promoting neuroinflammation and adult neurogenesis decline in hippocampus during SD, thereby, complement (C3aR/C5aR) antagonist may serve as the novel therapeutics to improve the SD mediated consequences.


Assuntos
Ativação do Complemento/imunologia , Neuroimunomodulação/fisiologia , Privação do Sono/metabolismo , Animais , Arginina/análogos & derivados , Arginina/farmacologia , Compostos Benzidrílicos/farmacologia , Disfunção Cognitiva/imunologia , Disfunção Cognitiva/metabolismo , Ativação do Complemento/fisiologia , Complemento C3a/metabolismo , Hipocampo/metabolismo , Masculino , Neurogênese/imunologia , Neurogênese/fisiologia , Neuroimunomodulação/imunologia , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Complemento/metabolismo , Transdução de Sinais/fisiologia , Privação do Sono/imunologia , Memória Espacial/fisiologia , Lobo Temporal/metabolismo
4.
Nanotoxicology ; 13(7): 977-989, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31012773

RESUMO

Aluminum oxide (Al2O3) nanoparticles (NPs) have a wide number of applications which cause intentional and unintentional exposure to humans, making it important to understand the nano-bio interaction. In this study, we made an attempt to evaluate the toxic effects of Al2O3 NPs chronic exposure on Drosophila melanogaster. Flies were exposed to Al2O3 NPs at concentration 0.1 and 1 mM via ingestion throughout their lifespan and progeny flies were screened for behavioral and phenotypic abnormalities. Behavioral abnormalities in flies were recorded through larval crawling, climbing in flies and two taste testing. Chronic exposure of Al2O3 NPs resulted in the loss of appendages in flies resulting in five legs flies, four legs flies and absence of haltere. Exposure to Al2O3 NPs caused renal failure in flies as observed by swollen abdomen. Our observations clearly showed that these NPs could cause detrimental health ailments which relate to human birth deformities and kidney failure. Damage at the cellular level was studied through proteomic profiling. Three hundred and seven unique proteins were expressed on exposure to Al2O3 NPs and 51 proteins were differentially expressed. Enrichment analysis of differentially expressed proteins showed significant alteration in striated muscle cell differentiation, digestive tract morphogenesis, phototransduction, regulation of chromatin organization and DNA duplex unwinding.


Assuntos
Óxido de Alumínio/toxicidade , Nanopartículas Metálicas/toxicidade , Animais , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/metabolismo , Humanos
5.
Physiol Behav ; 175: 56-65, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28351559

RESUMO

Hypobaric hypoxia leads to decrease in cellular oxygen content which subsequently damages the hippocampus with an increase in brain oxidative stress and impairs the memory of the individual. In the present study, we have evaluated the cognitive impairment modulating activity of total oligomeric flavonoids fraction of Cyperus rotundus (TOF) in Sprague Dawley rats. The rats were trained for memory activity for a period of 7days followed by 7days exposure to 25,000ft. altitude and the spatial reference memory was evaluated. Behavioral analysis of the rats by Morris water maze experiment showed that TOF supplementation enhanced the spatial reference memory activity of the rats exposed to hypobaric hypoxia. The decrease in antioxidant status of the animals exposed to hypoxia was restored with TOF supplementation. The increase in ROS, lipid peroxidation products and protein carbonyls of the hippocampus was significantly decreased in animals with TOF administration. The histological assessment of the pyramidal cells of the hippocampus of hypoxia-exposed animals showed nuclear damage and TOF supplementation prevented nuclear damage. TOF administration suppressed hypoxia-induced increase in serotonin, dopamine, and norepinephrine. GABA and Ach levels were decreased by hypoxia which was prevented by TOF supplementation. The increase in GFAP, HIF-1α and VEGF expression in CA3 region of the hippocampus in hypoxia-exposed rats was decreased in TOF administered rats. Taken together, TOF extract ameliorates hypobaric hypoxia induced memory impairment and neurodegeneration in hippocampus through its anti-stress effects.


Assuntos
Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Cyperus/química , Flavonoides/uso terapêutico , Hipóxia/complicações , Extratos Vegetais/uso terapêutico , Acetilcolinesterase/metabolismo , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Glutationa/metabolismo , L-Lactato Desidrogenase/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Rememoração Mental/efeitos dos fármacos , Neurotransmissores/metabolismo , Nitritos/metabolismo , Carbonilação Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
6.
Behav Brain Res ; 240: 76-86, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23159706

RESUMO

Chronic exposure to hypobaric hypoxia (HH) causes neurodegeneration and loss of memory. The underlying mechanisms of HH induced memory impairment have been attributed to prolonged elevated corticosterone level in hippocampus leading to augmented glutamate excitotoxicity, oxidative stress, alteration of neurotransmitter level or their receptors and calcium mediated signaling. Whether this corticosterone mediated neurodegenerative effect occurs through overstimulation of glucocorticoid receptors (GRs) or is independent of the GRs, is not known. Four groups of rats were taken and GR blocker mifepristone was administered intraperitoneally during exposure to HH from 3rd to 7th days. Our results showed a duration dependent transcriptional upregulation of GRs and MRs following exposure to HH. Prolonged exposure to HH for 7 days augmented the translocation of GRs from cytosol to nucleus. Inhibition of GRs during hypoxic exposure improved the hippocampal ATP level and modulated the apoptotic markers like p53, Bcl(2) and Bax. Decreased expression of L-type calcium channel and NR1 subunit of NMDA receptors were also observed following administration of mifepristone during hypoxic exposure. Morphological studies following mifepristone administration during hypoxic exposure showed decreased number of pyknotic cells in hippocampus and decrease in apoptotic and necrotic cells in the CA3 region of hippocampus. The study indicates that elevated corticosterone level during hypoxic exposure causes neurodegeneration and acts through its binding to GRs indicating that inhibition of GRs may provide therapeutic effect in ameliorating HH induced memory impairment.


Assuntos
Corticosterona/fisiologia , Hipóxia Encefálica/complicações , Transtornos da Memória/etiologia , Mifepristona/farmacologia , Receptores de Glucocorticoides/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Hipóxia Encefálica/metabolismo , Hipóxia Encefálica/patologia , Masculino , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Necrose , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Proteína Supressora de Tumor p53/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima , Proteína X Associada a bcl-2/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo
7.
J Ethnopharmacol ; 145(2): 431-41, 2013 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-23211660

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Withania somnifera (WS) root extract has been used traditionally in ayurvedic system of medicine as a memory enhancer and anti-stress agent. AIM OF THE STUDY: To evaluate the neuroprotective and prophylactic potential of WS root extract in ameliorating hypobaric hypoxia (HH) induced memory impairment and to explore the underlying molecular mechanism. MATERIALS AND METHODS: WS root extract was administered to male Sprague Dawley rats during a period of 21 days pre-exposure and 07 days exposure to a simulated altitude of 25,000 ft. Spatial memory was assessed by Morris Water Maze. Neurodegeneration, corticosterone, acetylcholine (Ach) levels, acetylcholine esterase (AchE) activity, oxidative stress markers and nitric oxide (NO) concentration were assessed in the hippocampus. Synaptic and apoptotic markers were also investigated by immunoblotting. To study the role of NO in regulating corticosterone mediated signaling, the neuronal nitric oxide synthase (n-NOS) inhibitor, L-Nitro-arginine methyl ester (L-Name) and NO agonist sodium nitroprusside (SNP) were administered from 3rd to 7th day of hypoxic exposure. RESULTS: Administration of WS root extract prevented HH induced memory impairment and neurodegeneration along with decreased NO, corticosterone, oxidative stress and AchE activity in hippocampal region. Inhibition of NO synthesis by administration of L-Name reduced corticosterone levels in hippocampus during hypoxic exposure while co-administration of corticosterone increased neurodegeneration. Administration of sodium nitroprusside (SNP) along with WS root extract supplementation during hypoxic exposure increased corticosterone levels and increased the number of pyknotic cells. CONCLUSION: WS root extract ameliorated HH induced memory impairment and neurodegeneration in hippocampus through NO mediated modulation of corticosterone levels.


Assuntos
Transtornos da Memória/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Fitoterapia , Extratos Vegetais/uso terapêutico , Withania , Acetilcolina/metabolismo , Acetilcolinesterase/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Canais de Cálcio Tipo L/metabolismo , Corticosterona/metabolismo , Glutationa/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipóxia/complicações , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/etiologia , Transtornos da Memória/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Extratos Vegetais/farmacologia , Raízes de Plantas , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Sinaptofisina/metabolismo
8.
Int J Biochem Cell Biol ; 44(1): 211-23, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22064248

RESUMO

Cellular potassium homeostasis has recently emerged as a critical regulator of apoptosis in response to variety of stimuli. However, functional hierarchy of this phenomenon in the apoptotic cascade and therefore, its significance as a pathway for intervention is not fully established. Chronic hypoxia, a known threat to cell survival, also modulates cellular potassium homeostasis. In this study, we tested if hypoxia-induced apoptosis in lymphocytes can be prevented by modulating cellular K+ homeostasis. We observed that chronic hypoxia accelerated the rate of apoptosis in resting murine splenocytes concomitant with cytosolic K+ efflux. We tested several modalities including elevated extracellular potassium besides various K+ channel inhibitors to curtail hypoxia-induced K+ efflux and interestingly, established that the supplementation of KCl in extracellular medium was most effective in preventing hypoxia-induced apoptosis in these cells. Subsequent mechanistic dissection of pathways underlying this phenomenon revealed that besides effectively inhibiting hypoxia-induced efflux of K+ ion and its downstream cell-physiological consequences; elevated extracellular KCl modulated steady state levels of cellular ATP and culminated in stabilization of AMPKα with pro-survival consequences. Also, interestingly, global gene expression profiling revealed that KCl supplementation down regulated a distinct p53-regulated cellular sub-network of genes involved in regulation of DNA replication. Additionally, we present experimental evidence for the functional role of AMPK and p53 activation during suppression of hypoxia-induced apoptosis. In conclusion, our study highlights a novel bimodal effect wherein cooperativity between restoration of K+ homeostasis and a sustainable 'metabolic quiescence' induced by AMPK activation appeared indispensible for curtailing hypoxia-induced apoptosis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Potássio/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Trifosfato de Adenosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Membrana Celular/metabolismo , Células Cultivadas , Citosol/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/metabolismo , Cloreto de Potássio/farmacologia , Fase S/efeitos dos fármacos , Fase S/fisiologia , Baço/citologia , Baço/efeitos dos fármacos , Baço/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
9.
Food Chem ; 127(3): 1309-16, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25214131

RESUMO

A novel environmentally friendly technique, subcritical water extraction (SWE) was employed for the extraction of antioxidant compounds from Seabuckthorn leaves (SBT). Antioxidant activity of the extracts was evaluated using commonly accepted chemical assays. Also, present study reports the cytoprotective and antioxidant properties of SBT against tertiary-butyl hydroperoxide (tert-BOOH) induced oxidative stress in murine macrophages (Raw 264.7). Exposure of cells to tert-BOOH resulted, increase in cytotoxicity, reactive oxygen species (ROS) production and decrease in mitochondrial membrane potential, which is responsible for fall in intracellular antioxidant levels. Pretreatment of cells with SBT extracts inhibited cytotoxicity, ROS production and maintained antioxidants levels similar to that of control cells. The chemical composition of the SWE extracts studied showed total phenol content (76.07-93.72mg/g GAE) and total flavonoid content (47.06-66.03mg/g rutin). Further, some of its phenolic constituents; (1) Quercetin-3-galactoside, (2) Kaempferol and (3) Isorhamnetin were quantified by RP-HPLC.

10.
Neurobiol Dis ; 34(1): 23-39, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19154788

RESUMO

Hypobaric hypoxia induced memory impairment has been attributed to several factors including increased oxidative stress, depleted mitochondrial bioenergetics, altered neurotransmission and apoptosis. This multifactorial response of the brain to hypobaric hypoxia limits the use of therapeutic agents that target individual pathways for ameliorating hypobaric hypoxia induced memory impairment. The present study aimed at exploring the therapeutic potential of a bacoside rich leaf extract of Bacopa monniera in improving the memory functions in hypobaric conditions. The learning ability was evaluated in male Sprague Dawley rats along with memory retrieval following exposure to hypobaric conditions simulating an altitude of 25,000 ft for different durations. The effect of bacoside administration on apoptosis, cytochrome c oxidase activity, ATP levels, and oxidative stress markers and on plasma corticosterone levels was investigated. Expression of NR1 subunit of N-methyl-d-aspartate receptors, neuronal cell adhesion molecules and was also studied along with CREB phosphorylation to elucidate the molecular mechanisms of bacoside action. Bacoside administration was seen to enhance learning ability in rats along with augmentation in memory retrieval and prevention of dendritic atrophy following hypoxic exposure. In addition, it decreased oxidative stress, plasma corticosterone levels and neuronal degeneration. Bacoside administration also increased cytochrome c oxidase activity along with a concomitant increase in ATP levels. Hence, administration of bacosides could be a useful therapeutic strategy in ameliorating hypobaric hypoxia induced cognitive dysfunctions and other related neurological disorders.


Assuntos
Bacopa , Hipóxia/fisiopatologia , Transtornos da Memória/tratamento farmacológico , Fitoterapia , Extratos Vegetais/uso terapêutico , Trifosfato de Adenosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Moléculas de Adesão Celular Neuronais/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Masculino , Fosforilação/efeitos dos fármacos , Extratos Vegetais/administração & dosagem , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Percepção Espacial/efeitos dos fármacos
11.
J Neurosci Res ; 86(12): 2705-21, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18500755

RESUMO

Cellular and molecular pathways underlying hypoxic neurotoxicity and cell death are multifaceted and complex. Although many potentially neuroprotective agents have been investigated, the protection conferred is often inadequate, resulting in their insufficient clinical utility. In light of the above, we investigated the therapeutic potential and mechanism of action of acetyl-L-carnitine (ALCAR) in protecting hippocampal neurons from hypoxia-induced neurotoxicity and cellular death. Results showed decreased viability of hippocampal cells when exposed to hypoxia (3% O(2)) for 48 hr along with concomitant membrane depolarization, adenosine triphosphate depletion, DNA fragmentation, accentuated free radical production, and lactate dehydrogenase activity. Pretreatment with ALCAR significantly attenuated hypoxia-induced cytotoxicity in a dose-dependent manner and improved cellular glutathione levels and cytochrome c oxidase activity compared with normoxic controls. Supplementation of ALCAR also prevented apoptosis by down-regulating caspase-3 levels, cytochrome c release, and p-Bcl-2 expression. A decrease in nerve growth factor (NGF) was observed in hypoxic stress despite increased phosphorylation of ERK1/2 (extracellular signal-related kinase) and its downstream effector, Elk-1. Supplementation of ALCAR, on the other hand, up-regulated NGF and tyrosine kinase A expression along with concomitant increase in ERK1/2 phosphorylation, thus enhancing cell survival. ALCAR therefore provides neuroprotection by stabilizing mitochondrial membrane, restoring the cholinergic transmission, and more importantly, it stimulates NGF receptors, thus triggering cell survival pathway via ERK phosphorylation. Therefore, ALCAR may be useful as an effective therapeutic agent for hypoxic stress and associated neurodegenerative diseases.


Assuntos
Acetilcarnitina/farmacologia , Hipocampo/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fator de Crescimento Neural/metabolismo , Fármacos Neuroprotetores/farmacologia , Animais , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Células Cultivadas , Hipocampo/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Fator de Crescimento Neural/genética , Ratos , Ratos Sprague-Dawley
12.
Mol Cell Neurosci ; 33(1): 96-108, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16901715

RESUMO

The TAM receptor tyrosine kinase Mer is expressed by cells of the retinal pigment epithelium (RPE), and genetic studies have demonstrated that Mer is essential for RPE function. RPE cells that lack Mer exhibit a severely compromised ability to phagocytose the distal ends of photoreceptor (PR) outer segments, which leads to the complete postnatal degeneration of photoreceptors and to blindness. Although in vitro experiments have implicated Gas6 as the critical TAM ligand for this process, we find that Gas6 mutant mice have a histologically intact retina with no photoreceptor degeneration. We further find that, in addition to Mer, RPE cells also express another TAM receptor--Tyro 3--and that both of these receptors are instead activated independently by the Gas6-related ligand Protein S. This protein is also expressed by RPE cells. Finally, we demonstrate that loss of Mer function is accompanied by a substantial down-regulation in Tyro 3 as well. These observations indicate that both Mer and Tyro 3 act in mouse RPE cells and suggest that their biologically relevant ligand in these cells is Protein S.


Assuntos
Epitélio Pigmentado Ocular , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Células Cultivadas , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Ligantes , Camundongos , Camundongos Knockout , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/ultraestrutura , Epitélio Pigmentado Ocular/citologia , Epitélio Pigmentado Ocular/crescimento & desenvolvimento , Epitélio Pigmentado Ocular/metabolismo , Proteína S/metabolismo , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , c-Mer Tirosina Quinase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA