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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Med ; 17(5-6): 448-56, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21274503

RESUMO

Histone deacetylase (HDAC) inhibitors are emerging as a novel class of potentially therapeutic agents for treating acute injuries of the central nervous system (CNS). In this review, we summarize data regarding the effects of HDAC inhibitor administration in models of acute CNS injury and discuss issues warranting clinical trials. We have previously shown that the pan-HDAC inhibitor ITF2357, a compound shown to be safe and effective in humans, improves functional recovery and attenuates tissue damage when administered as late as 24 h after injury. Using a well-characterized, clinically relevant mouse model of closed head injury, we demonstrated that a single dose of ITF2357 administered 24 h after injury improves neurobehavioral recovery and reduces tissue damage. ITF2357-induced functional improvement was found to be sustained up to 14 d after trauma and was associated with augmented histone acetylation. Single postinjury administration of ITF2357 also attenuated injury-induced inflammatory responses, as indicated by reduced glial accumulation and activation as well as enhanced caspase-3 expression within microglia/macrophages after treatment. Because no specific therapeutic intervention is currently available for treating brain trauma patients, the ability to affect functional outcome by postinjury administration of HDAC inhibitors within a clinically feasible timeframe may be of great importance. Furthermore, a growing body of evidence indicates that HDAC inhibitors are beneficial for treating various forms of acute CNS injury including ischemic and hemorrhagic stroke. Because HDAC inhibitors are currently approved for other use, they represent a promising new avenue of treatment, and their use in the setting of CNS injury warrants clinical evaluation.


Assuntos
Sistema Nervoso Central/lesões , Sistema Nervoso Central/patologia , Inibidores de Histona Desacetilases/uso terapêutico , Traumatismos do Sistema Nervoso/tratamento farmacológico , Animais , Sistema Nervoso Central/efeitos dos fármacos , Humanos , Ácidos Hidroxâmicos/uso terapêutico , Camundongos , Traumatismos do Sistema Nervoso/metabolismo
2.
FASEB J ; 23(12): 4266-75, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19723705

RESUMO

Despite efforts aimed at developing novel therapeutics for traumatic brain injury (TBI), no specific pharmacological agent is currently clinically available. Here, we show that the pan-histone deacetylase (HDAC) inhibitor ITF2357, a compound shown to be safe and effective in humans, improves functional recovery and attenuates tissue damage when administered as late as 24 h postinjury. Using a well-characterized, clinically relevant mouse model of closed head injury (CHI), we demonstrate that a single dose of ITF2357 administered 24 h postinjury improves neurobehavioral recovery from d 6 up to 14 d postinjury (improved neurological score vs. vehicle; P< or =0.05), and that this functional benefit is accompanied by decreased neuronal degeneration, reduced lesion volume (22% reduction vs. vehicle; P< or =0.01), and is preceded by increased acetylated histone H3 levels and attenuation of injury-induced decreases in cytoprotective heat-shock protein 70 kDa and phosphorylated Akt. Moreover, reduced glial accumulation and activation were observed 3 d postinjury, and total p53 levels at the area of injury and caspase-3 immunoreactivity within microglia/macrophages at the trauma area were elevated, suggesting enhanced clearance of these cells via apoptosis following treatment. Hence, our findings underscore the relevance of HDAC inhibitors for ameliorating trauma-induced functional deficits and warrant consideration of applying ITF2357 for this indication.


Assuntos
Apoptose/efeitos dos fármacos , Lesões Encefálicas/tratamento farmacológico , Histona Desacetilases/metabolismo , Ácidos Hidroxâmicos/farmacologia , Neuroglia/efeitos dos fármacos , Animais , Encéfalo/patologia , Caspase 3/metabolismo , Ácidos Hidroxâmicos/administração & dosagem , Masculino , Camundongos , Fármacos Neuroprotetores/farmacologia
3.
J Neurotrauma ; 25(2): 112-23, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18260794

RESUMO

Both heat acclimation (HA) and post-injury treatment with recombinant human erythropoietin (Epo, rhEpo, exogenous Epo) are neuroprotective against traumatic brain injury (TBI). Our previous data demonstrated that HA-induced neuroprotection includes improved functional recovery and reduced cerebral edema formation. Additionally, in earlier Western-blot analyses, we found that HA mice display increased expression of the specific erythropoietin receptor (EpoR) and of hypoxia-inducible factor-1 alpha (HIF-1 alpha), the inducible subunit of the transcription factor, which regulates Epo gene expression, but not of Epo itself. In light of this, the aim of the current study was threefold: (1) to assess Epo expression in the trauma area and hippocampus following HA, rhEpo administration, or combined HA-rhEpo treatment, using immunohistochemical methods that offer enhanced anatomical resolution; (2) to examine the effects of endogenous and exogenous Epo on edema formation in normothermic (NT) mice; and (3) to evaluate the effects of exogenous Epo administration on neuroprotective outcome measures in HA animals. HA induced enhanced expression of endogenous Epo in the trauma area and the hippocampus. Treatment with anti-Epo antibody given to NT mice increased edema formation, whereas rhEpo induced no beneficial effect. Cognitive performance testing and immunohistochemical findings reinforced HA and rhEpo as separate protective interventions but showed no advantage to combining the two strategies. We therefore suggest that HA-induced neuroprotection is shaped by pre-existing mediators but cannot be modified by post-injury treatment aimed at increasing the levels of neuroprotective agents.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Eritropoetina/farmacologia , Eritropoetina/fisiologia , Fármacos Neuroprotetores , Aclimatação , Animais , Edema Encefálico/etiologia , Edema Encefálico/prevenção & controle , Lesões Encefálicas/patologia , Cognição/fisiologia , Eritropoetina/biossíntese , Eritropoetina/uso terapêutico , Fluoresceínas , Corantes Fluorescentes , Hipocampo/patologia , Temperatura Alta , Humanos , Imuno-Histoquímica , Imunoterapia , Masculino , Camundongos , Degeneração Neural/patologia , Compostos Orgânicos , Reconhecimento Psicológico/fisiologia , Proteínas Recombinantes
4.
J Neurochem ; 103(4): 1523-9, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17725578

RESUMO

Long-term heat exposure, known as heat acclimation (HA; 30 days at 34 +/- 1 degrees C) is neuroprotective against traumatic brain injury. Acclimated mice were previously found to display improved functional recovery as well as an increase in the levels of the specific erythropoietin receptor. As the activation of this receptor is known to facilitate functional recovery on one hand and the phosphorylation and activation of Akt, an intracellular kinase which regulates anti-apoptotic pathways on the other, in this study we investigated whether HA affects Akt phosphorylation prior to and following injury and whether this step is required for development of HA-induced neuroprotection. Akt phosphorylation was blocked using Triciribine (TCN), a compound shown to block the phosphorylation process without affecting upstream effectors of this kinase, and several post-injury functional end-point measures were subsequently evaluated. Acclimation led to a post-injury increase in the levels of phosphorylated Akt, resulting in higher levels when compared with normothermic controls at 4 h post-injury (63.6 +/- 5.2% and 42.7 +/- 3.7%, respectively, p

Assuntos
Aclimatação/fisiologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/prevenção & controle , Temperatura Alta/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Temperatura Corporal/fisiologia , Lesões Encefálicas/fisiopatologia , Masculino , Camundongos , Fármacos Neuroprotetores/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Fatores de Tempo
5.
Prog Brain Res ; 161: 353-63, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17618990

RESUMO

Sub-lethal exposure to practically any harmful stimulus has been shown to induce consequent protection against more severe stress. This preconditioning (PC) effect may be achieved by exposure to different stressors, indicating that the induction of tolerance involves activation of common protective pathways. Chronic exposure to moderate heat (heat acclimation, HA) is a unique PC model, since this global physiological adaptation, as opposed to discrete organ PC, has been shown to induce cross-tolerance against other stressors, including closed head injury (CHI). HA animals show accelerated functional recovery after injury which is accompanied by reduced secondary brain damage. However, the precise mechanisms underlying this phenomenon have not been thoroughly studied until recently. Here we will address the concept of PC, highlighting the unique properties of HA as a model which can be used for the study of endogenous protective pathways triggered by PC procedures. Several molecular mechanisms which are suggested to mediate HA-induced neuroprotection will also be discussed, bringing to light their potential contribution to the development of traumatic brain injury treatment strategies utilizing therapeutic augmentation of endogenous defense mechanisms.


Assuntos
Aclimatação/fisiologia , Lesões Encefálicas/fisiopatologia , Temperatura Alta , Animais , Antioxidantes/metabolismo , Humanos
6.
Brain Res ; 1185: 313-20, 2007 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-17963735

RESUMO

Long-term exposure to moderate ambient heat (heat acclimation, HA, 30 days at 34+/-1 degrees C) provides protection toward a variety of stressors including traumatic brain injury. As previous studies suggested an anti-inflammatory effect of HA and given the ability of augmented pre-injury anti-inflammatory cytokine expression to harbor neuroprotection and to attenuate early post-injury expression of pro-inflammatory mediators, we hypothesized that HA-induced neuroprotection may involve enhanced pre-injury expression of anti-inflammatory mediators or a reduction in post-injury TNF alpha (TNFalpha) expression. Since the attenuation of inflammatory-associated entities has also been linked to mild hypothermia, an established neuroprotective paradigm, the effect of HA on post-injury body temperature was also studied. HA mice and normothermic (NT) counterparts were examined using a closed head injury model. Cytokines were measured within the ipsilateral cortex. Pre-injury protein levels of anti-inflammatory interleukins 10 and 4 (IL-10, IL-4) were quantified by enzyme-linked immunosorbent assays (ELISA). mRNA and protein levels of TNFalpha were quantified during the initial 2 h post-injury using semi-quantitative and real-time polymerase chain reaction (sqRT-PCR and qRT-PCR) or ELISA, respectively. Rectal temperatures were measured. HA induced augmented pre-injury IL-10 expression and a post-injury reduction in TNFalpha mRNA levels, as well as altered expression dynamics of TNFalpha protein. TNFalpha protein levels decreased relative to the sham state in HA mice only. HA mice displayed sustained post-injury hypothermia, namely significantly lower body temperature at 4 h post-injury. Given the evidence on the neuroprotective nature of hypothermia and anti-inflammatory cytokines, we suggest that these changes may contribute to HA-induced neuroprotection.


Assuntos
Aclimatação/fisiologia , Lesões Encefálicas/complicações , Citocinas/metabolismo , Regulação da Expressão Gênica/fisiologia , Temperatura Alta , Hipotermia/etiologia , Análise de Variância , Animais , Ensaio de Imunoadsorção Enzimática/métodos , Masculino , Camundongos , Fatores de Tempo
7.
J Cereb Blood Flow Metab ; 25(11): 1456-65, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15902197

RESUMO

Experimental evidence indicates that long-term exposure to moderately high ambient temperature (heat acclimation, HA) mediates cross-tolerance to various types of subsequently applied stress. The transcriptional activator hypoxia-inducible factor 1 (HIF-1) has been implicated in playing a critical role in HA. It also regulates the expression of Erythropoietin (Epo), whose neuroprotective effects have been shown in a variety of brain injuries. The aim of the present study was to examine whether HA exerts a beneficial effect on the outcome of closed head injury (CHI) in mice and to explore the possible involvement of HIF-1 and Epo in this process. Heat acclimated mice and matched normothermic controls were subjected to CHI or sham surgery. Postinjury motor and cognitive parameters of acclimated mice were compared with those of controls. Mice were killed at various time points after injury or sham surgery and brain levels of HIF-1alpha, the inducible subunit of HIF-1, Epo, and the specific erythropoietin receptor (EpoR) were analyzed by Western immunoblotting. Motor and cognitive functions of acclimated mice were significantly better than those of controls. Heat acclimation was found to induce a significant increase in expression of nuclear HIF-1alpha and EpoR. The EpoR/Epo ratio was also significantly higher in acclimated mice as compared with controls. Nuclear HIF-1alpha and EpoR were higher in the acclimated group at 4 h after injury as well. The improved outcome of acclimated mice taken together with the basal and postinjury upregulation of the examined proteins suggests the involvement of this pathway in HA-induced neuroprotection.


Assuntos
Aclimatação , Lesões Encefálicas/metabolismo , Temperatura Alta , Receptores da Eritropoetina/biossíntese , Regulação para Cima , Animais , Química Encefálica , Lesões Encefálicas/patologia , Cognição , Eritropoetina/biossíntese , Masculino , Camundongos , Atividade Motora , Receptores da Eritropoetina/genética
8.
J Cereb Blood Flow Metab ; 30(3): 616-27, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19904288

RESUMO

Heat acclimation (HA) offers functional neuroprotection in mice after traumatic brain injury (TBI). This study further characterizes endogenous neuroprotection acquired by HA (34+/-1 degrees C, 30 d) after TBI. We establish here the ability of HA to induce sustained functional benefits and to reduce activation of apoptotic pathways. Neurobehavioral recovery, assessed by the Neurological Severity Score, was greater in HA mice up to 8 days after injury as compared with normothermic controls (P<0.05) and lesion volume was also smaller in the HA group (P<0.05). Reduced apoptotic cell death in HA mice was confirmed using caspase-3 activity measurements and immunohistochemistry. To investigate the underlying molecular pathways, expression levels of intrinsic apoptotic pathway-related proteins were examined. HA mice displayed higher mitochondrial levels of antiapoptotic Bcl-xL, accompanied by lower proapoptotic Bad levels and decreased cytochrome c release, suggesting a higher apoptotic threshold. Taken together with our previous reports, indicating increased Akt phosphorylation and antioxidative capacity, alongside with reduced tumor necrosis alpha levels after TBI in HA animals, the current results support the involvement of an antiapoptotic effect in HA-induced neuroprotection. Current results warrant further study as TBI-induced apoptosis may persist over weeks after injury, possibly providing a target for belated therapeutic intervention.


Assuntos
Aclimatação/fisiologia , Apoptose/fisiologia , Lesões Encefálicas/patologia , Temperatura Alta , Recuperação de Função Fisiológica/fisiologia , Animais , Antioxidantes/metabolismo , Comportamento Animal/fisiologia , Western Blotting , Lesões Encefálicas/fisiopatologia , Caspase 3/metabolismo , Citocromos c/biossíntese , Coração/fisiopatologia , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Miocárdio/metabolismo , Miocárdio/patologia , Estresse Oxidativo/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína de Morte Celular Associada a bcl/biossíntese , Proteína bcl-X/biossíntese
10.
Brain Res ; 1244: 132-41, 2008 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-18835383

RESUMO

Brain-derived neurotrophic factor (BDNF) conveys neuroprotection in various settings of experimental central nervous system injury. Using a model of endogenous neuroprotection, induced in mice by chronic exposure to moderate ambient heat (heat acclimation, HA), we have previously shown that neuroprotection following traumatic brain injury involves reduced post-injury tumor necrosis factor alpha (TNFalpha) expression. As glial cells play a pivotal role in post-injury inflammation on one hand, and are also capable of inducing neuroprotection by harboring trophic factors and BDNF in particular, the effects of injury and HA on overall BDNF content at the trauma area, gliosis and glial BDNF expression were investigated. Western blotting indicated higher overall BDNF levels in HA sham-operated mice. Following injury, a decrease was observed in the HA group only, reaching levels similar to normothermic mice. Immunohistochemical studies demonstrated BDNF-positive resting microglia in non-injured HA but not normothermic animals. Post-injury astrocytosis and microglial immunoreactivity were enhanced in the HA group. Particularly, an increase in the amount of ramified microglia was observed within the penumbra, accompanied by a concomitant decrease in globular microglia, a major source of pro-inflammatory mediators. BDNF labeling on and around microglia and their processes was intensified in HA mice. Furthermore, BDNF immunoreactivity in HA mice was evident in the degenerated edges of axons. These findings, taken together with the growing body of evidence indicating the neuroprotective potential of both BDNF and microglia, suggest a possible role of these cells in HA-induced neuroprotection.


Assuntos
Aclimatação/fisiologia , Lesões Encefálicas/fisiopatologia , Temperatura Alta , Microglia/fisiologia , Análise de Variância , Animais , Astrócitos/metabolismo , Astrócitos/fisiologia , Western Blotting , Lesões Encefálicas/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/metabolismo , Gliose/fisiopatologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos , Microglia/metabolismo
11.
J Cereb Blood Flow Metab ; 28(7): 1341-52, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18414499

RESUMO

Boswellia resin has been used as a major anti-inflammatory agent and for the healing of wounds for centuries. Incensole acetate (IA), isolated from this resin, was shown to inhibit the activation of nuclear factor-kappaB, a key transcription factor in the inflammatory response. We now show that IA inhibits the production of inflammatory mediators in an in vitro model system of C6 glioma and human peripheral monocytes. Given the involvement of postinjury inflammation in the pathophysiology and outcome of traumatic brain injury, we examined the effect of IA on the inflammatory process and on the recovery of neurobehavioral and cognitive functions in a mouse model of closed head injury (CHI). In the brains of post-CHI mice, IA reduced glial activation, inhibited the expression of interleukin-1beta, and tumor necrosis factor-alpha mRNAs, and induced cell death in macrophages at the area of trauma. A mild hypothermic effect was also noted. Subsequently, IA inhibited hippocampal neurodegeneration and exerted a beneficial effect on functional outcome after CHI, indicated by reduced neurological severity scores and improved cognitive ability in an object recognition test. This study attributes the anti-inflammatory activity of Boswellia resin to IA and related cembranoid diterpenes and suggests that they may serve as novel neuroprotective agents.


Assuntos
Boswellia/química , Lesões Encefálicas/tratamento farmacológico , Diterpenos/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Lesões Encefálicas/patologia , Citocinas/efeitos dos fármacos , Diterpenos/isolamento & purificação , Diterpenos/uso terapêutico , Hipocampo/patologia , Mediadores da Inflamação/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , Camundongos , Modelos Animais , Fármacos Neuroprotetores/isolamento & purificação , Fármacos Neuroprotetores/uso terapêutico , Recuperação de Função Fisiológica/efeitos dos fármacos
12.
Neurobiol Dis ; 22(2): 257-64, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16364651

RESUMO

Endocannabinoids are involved in neuroprotection through numerous biochemical pathways. We have shown that the endocannabinoid 2-arachidonoyl glycerol (2-AG) is released in mouse brain after closed head injury (CHI), and treatment with exogenous 2-AG exerts neuroprotection via the central cannabinoid receptor CB1. This process involves inhibition of inflammatory signals that are mediated by activation of the transcription factor NF-kB. The present study was designed to examine the effect of 2-AG on the blood-brain barrier (BBB) and the possible inhibition of the early expression of proinflammatory cytokines, which are implicated in BBB disruption. We found that 2-AG decreased BBB permeability and inhibited the acute expression of the main proinflammatory cytokines: TNF-alpha, IL-1beta and IL-6. It also augmented the levels of endogenous antioxidants. We suggest that 2-AG exerts neuroprotection in part by inhibition of the early (1-4 h) inflammatory response and augmentation of the brain reducing power.


Assuntos
Ácidos Araquidônicos/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Citocinas/antagonistas & inibidores , Encefalite/tratamento farmacológico , Glicerídeos/farmacologia , Traumatismos Cranianos Fechados/complicações , Fármacos Neuroprotetores/farmacologia , Animais , Antioxidantes/metabolismo , Ácidos Araquidônicos/metabolismo , Ácidos Araquidônicos/uso terapêutico , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/fisiopatologia , Citocinas/genética , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Encefalite/etiologia , Encefalite/fisiopatologia , Endocanabinoides , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Glicerídeos/metabolismo , Glicerídeos/uso terapêutico , Traumatismos Cranianos Fechados/fisiopatologia , Interleucina-1/antagonistas & inibidores , Interleucina-1/genética , Interleucina-6/antagonistas & inibidores , Interleucina-6/genética , Masculino , Camundongos , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/uso terapêutico , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA