Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Neurochem ; 123(6): 1019-29, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23057870

RESUMO

Traumatic brain injury (TBI) results in significant inflammation which contributes to the evolving pathology. Previously, we have demonstrated that cyclic AMP (cAMP), a molecule involved in inflammation, is down-regulated after TBI. To determine the mechanism by which cAMP is down-regulated after TBI, we determined whether TBI induces changes in phosphodiesterase (PDE) expression. Adult male Sprague Dawley rats received moderate parasagittal fluid-percussion brain injury (FPI) or sham injury, and the ipsilateral, parietal cortex was analyzed by western blotting. In the ipsilateral parietal cortex, expression of PDE1A, PDE4B2, and PDE4D2, significantly increased from 30 min to 24 h post-injury. PDE10A significantly increased at 6 and 24 h after TBI. Phosphorylation of PDE4A significantly increased from 6 h to 7 days post-injury. In contrast, PDE1B, PD4A5, and PDE4A8 significantly decreased after TBI. No changes were observed with PDE1C, PDE3A, PDE4B1/3, PDE4B4, PDE4D3, PDE4D4, PDE8A, or PDE8B. Co-localization studies showed that PDE1A, PDE4B2, and phospho-PDE4A were neuronally expressed, whereas PDE4D2 was expressed in neither neurons nor glia. These findings suggest that therapies to reduce inflammation after TBI could be facilitated with targeted therapies, in particular for PDE1A, PDE4B2, PDE4D2, or PDE10A.


Assuntos
Lesões Encefálicas/enzimologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 1/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Regulação Enzimológica da Expressão Gênica/genética , Diester Fosfórico Hidrolases/genética , Animais , Lesões Encefálicas/genética , Lesões Encefálicas/terapia , AMP Cíclico/antagonistas & inibidores , AMP Cíclico/biossíntese , Nucleotídeo Cíclico Fosfodiesterase do Tipo 1/antagonistas & inibidores , Nucleotídeo Cíclico Fosfodiesterase do Tipo 1/biossíntese , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/biossíntese , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/biossíntese , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Modelos Animais de Doenças , Isoenzimas/antagonistas & inibidores , Isoenzimas/biossíntese , Isoenzimas/genética , Masculino , Diester Fosfórico Hidrolases/biossíntese , Diester Fosfórico Hidrolases/metabolismo , Fosforilação/genética , Ratos , Ratos Sprague-Dawley
2.
Eur J Neurosci ; 32(11): 1912-20, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21044182

RESUMO

Traumatic brain injury (TBI) is a major risk factor for the subsequent development of epilepsy. Currently, chronic seizures after brain injury are often poorly controlled by available antiepileptic drugs. Hypothermia treatment, a modest reduction in brain temperature, reduces inflammation, activates pro-survival signaling pathways, and improves cognitive outcome after TBI. Given the well-known effect of therapeutic hypothermia to ameliorate pathological changes in the brain after TBI, we hypothesized that hypothermia therapy may attenuate the development of post-traumatic epilepsy and some of the pathomechanisms that underlie seizure formation. To test this hypothesis, adult male Sprague Dawley rats received moderate parasagittal fluid-percussion brain injury, and were then maintained at normothermic or moderate hypothermic temperatures for 4 h. At 12 weeks after recovery, seizure susceptibility was assessed by challenging the animals with pentylenetetrazole, a GABA(A) receptor antagonist. Pentylenetetrazole elicited a significant increase in seizure frequency in TBI normothermic animals as compared with sham surgery animals and this was significantly reduced in TBI hypothermic animals. Early hypothermia treatment did not rescue chronic dentate hilar neuronal loss nor did it improve loss of doublecortin-labeled cells in the dentate gyrus post-seizures. However, mossy fiber sprouting was significantly attenuated by hypothermia therapy. These findings demonstrate that reductions in seizure susceptibility after TBI are improved with post-traumatic hypothermia and provide a new therapeutic avenue for the treatment of post-traumatic epilepsy.


Assuntos
Lesões Encefálicas/complicações , Epilepsia Pós-Traumática/etiologia , Epilepsia Pós-Traumática/terapia , Hipotermia Induzida , Animais , Temperatura Corporal , Proteína Duplacortina , Hipocampo/citologia , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Ratos , Ratos Sprague-Dawley
3.
J Cereb Blood Flow Metab ; 27(5): 939-49, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-16955078

RESUMO

In response to traumatic brain injury (TBI), neurons initiate neuroplastic processes through the activation of intracellular signaling pathways. However, the molecular mechanisms underlying neuroplasticity after TBI are poorly understood. To study this, we utilized the fluid-percussion brain injury (FPI) model to investigate alterations in the mammalian target of rapamycin (mTOR) signaling pathways in response to TBI. Mammalian target of rapamycin stimulates mRNA translation through phosphorylation of eukaryotic initiation factor 4E binding protein-1 (4E-BP1), p70 ribosomal S6 kinase (p70S6K), and ribosomal protein S6 (rpS6). These pathways coordinate cell growth and neuroplasticity via dendritic protein synthesis. Rats received sham surgery or moderate parasagittal FPI on the right side of the parietal cortex, followed by 15 mins, 30 mins, 4 h, 24 h, or 72 h of recovery. Using Western blot analysis, we found that mTOR, p70S6K, rpS6, and 4E-BP1 phosphorylation levels were significantly increased in the ipsilateral parietal cortex and hippocampus from 30 mins to 24 h after TBI, whereas total protein levels were unchanged. Using confocal microscopy to localize these changes, we found that rpS6 phosphorylation was increased in the parietal cortex and all subregions of the hippocampus. In accordance with these results, eIF4E, a key, rate-limiting mRNA translation factor, was also phosphorylated by mitogen-activated protein kinase-interacting kinase 1 (Mnk1) 15 mins after TBI. Together, these results suggest that changes in mRNA translation may be one mechanism that neurons use to respond to trauma and may contribute to the neuroplastic changes observed after TBI.


Assuntos
Lesões Encefálicas/fisiopatologia , Dendritos/patologia , Hipocampo/patologia , Plasticidade Neuronal/fisiologia , Proteínas Quinases/genética , Transdução de Sinais/fisiologia , Sinapses/patologia , Animais , Western Blotting , Proteínas de Transporte/metabolismo , Dendritos/ultraestrutura , Hipocampo/ultraestrutura , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Microscopia Confocal , Fosfoproteínas/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Proteínas Quinases S6 Ribossômicas/biossíntese , Proteínas Quinases S6 Ribossômicas/genética , Frações Subcelulares/metabolismo , Serina-Treonina Quinases TOR
4.
J Neurotrauma ; 24(4): 599-612, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17439344

RESUMO

Traumatic brain injury (TBI) initiates a complex genetic response that may include the expression of organelle specific stress genes. We investigated the effects of brain trauma on the expression of a number of stress genes by in situ hybridization and Western blot analysis including the endoplasmic reticulum (ER) stress gene grp78, ER protein processing enzymes calnexin and protein disulphide isomerase (PDI), the mitochondrial stress gene hsp60, and the cytoplasmic stress gene hsp70. Male Sprague-Dawley rats were subjected either to sham-surgery or moderate (1.8-2.2 atm) parasagittal fluid-percussion (F-P) brain injury followed by 30 min of either normoxic or hypoxic (30-40 mm Hg) gas levels. Expression of grp78 was increased in the ipsilateral cerebral cortex and dentate gyrus beginning 4 h after trauma plus hypoxia. Similarly, mRNA encoding the mitochondrial hsp60 was induced in the ipsilateral outer cortical layers at 4-24 h after TBI plus hypoxia. Calnexin and PDI mRNAs were not significantly altered following TBI with or without secondary hypoxia. In contrast, mRNA of the cytoplasmic hsp70 was strongly induced at 4 h after brain injury in multiple brain regions within the injured hemisphere, and this expression was greatly enhanced by secondary hypoxia. Because subcellular stress gene expression may reflect where unfolded or damaged proteins are abundant, these findings suggest that abnormal proteins are localized mainly in the cytoplasm, and to a lesser degree in the ER lumen and mitochondria after brain trauma. Thus, distinct parts of the cellular machinery respond to traumatic and metabolic stresses in specific ways.


Assuntos
Lesões Encefálicas/metabolismo , Frações Subcelulares/metabolismo , Animais , Western Blotting , Lesões Encefálicas/genética , Calnexina/biossíntese , Calnexina/genética , Chaperonina 60/biossíntese , Chaperonina 60/genética , Proteínas de Choque Térmico HSP70/biossíntese , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico/biossíntese , Proteínas de Choque Térmico/genética , Hibridização In Situ , Masculino , Chaperonas Moleculares/biossíntese , Chaperonas Moleculares/genética , Proteínas do Tecido Nervoso/biossíntese , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley
5.
J Cereb Blood Flow Metab ; 26(12): 1507-18, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16570077

RESUMO

A prominent cognitive impairment after traumatic brain injury (TBI) is hippocampal-dependent memory loss. Although the histopathologic changes in the brain are well documented after TBI, the underlying biochemical mechanisms that contribute to memory loss have yet to be thoroughly delineated. Thus, we determined if calcium/calmodulin-dependent protein kinases (CaMKs), known to be necessary for the formation of hippocampal-dependent memories, are regulated after TBI. Sprague-Dawley rats underwent moderate parasagittal fluid-percussion brain injury on the right side of the parietal cortex. The ipsilateral hippocampus and parietal cortex were Western blotted for phosphorylated, activated alpha-calcium/calmodulin-dependent protein kinase II (alpha-CaMKII), CaMKIV, and CaMKI. alpha-Calcium/calmodulin-dependent protein kinase II was activated in membrane subcellular fractions from the hippocampus and parietal cortex 30 mins after TBI. CaMKI and CaMKIV were activated in a more delayed manner, increasing in phosphorylation 1 h after TBI. The increase in activated alpha-CaMKII in membrane fractions was accompanied by a decrease in cytosolic total alpha-CaMKII, suggesting redistribution to the membrane. Using confocal microscopy, we observed that alpha-CaMKII was activated within hippocampal neurons of the dentate gyrus, CA3, and CA1 regions. Two downstream substrates of alpha-CaMKII, the AMPA-type glutamate receptor GluR1, and cytoplasmic polyadenylation element-binding protein, concomitantly increased in phosphorylation in the hippocampus and cortex 1 h after TBI. These results demonstrate that several of the biochemical cascades that subserve memory formation are activated unselectively in neurons after TBI. As memory formation requires activation of CaMKII signaling pathways at specific neuronal synapses, unselective activation of CaMKII signaling in all synapses may disrupt the machinery for memory formation, resulting in memory loss after TBI.


Assuntos
Lesões Encefálicas/enzimologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Córtex Cerebelar/enzimologia , Hipocampo/enzimologia , Transtornos da Memória/enzimologia , Memória , Animais , Lesões Encefálicas/patologia , Córtex Cerebelar/patologia , Ativação Enzimática , Hipocampo/patologia , Masculino , Transtornos da Memória/patologia , Neurônios/enzimologia , Neurônios/patologia , Fosforilação , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Sinapses/enzimologia , Sinapses/patologia , Fatores de Tempo
6.
J Neurosci ; 24(49): 11010-6, 2004 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-15590916

RESUMO

The tumor necrosis factor (TNF) ligand-receptor system plays an essential role in apoptosis that contributes to secondary damage after traumatic brain injury (TBI). TNF also stimulates inflammation by activation of gene transcription through the IkappaB kinase (IKK)/NF-kappaB and JNK (c-Jun N-terminal protein kinase)/AP-1 signaling cascades. The mechanism by which TNF signals between cell death and survival and the role of receptor localization in the activation of downstream signaling events are not fully understood. Here, TNF receptor 1 (TNFR1) signaling complexes in lipid rafts were investigated in the cerebral cortex of adult male Sprague Dawley rats subjected to moderate (1.8-2.2 atmospheres) fluid-percussion TBI and naive controls. In the normal rat cortex, a portion of TNFR1 was present in lipid raft microdomains, where it associated with the adaptor proteins TRADD (TNF receptor-associated death domain), TNF receptor-associated factor-2 (TRAF-2), the Ser/Thr kinase RIP (receptor-interacting protein), TRAF1, and cIAP-1 (cellular inhibitor of apoptosis protein-1), forming a survival signaling complex. Moderate TBI resulted in rapid recruitment of TNFR1, but not TNFR2 or Fas, to lipid rafts and induced alterations in the composition of signaling intermediates. TNFR1 and TRAF1 were polyubiquitinated in lipid rafts after TBI. Subsequently, the signaling complex contained activated caspase-8, thus initiating apoptosis. In addition, TBI caused a transient activation of NF-kappaB, but receptor signaling interacting proteins IKKalpha and IKKbeta were not detected in raft-containing fractions. Thus, redistribution of TNFR1 in lipid rafts and nonraft regions of the plasma membrane may regulate the diversity of signaling responses initiated by these receptors in the normal brain and after TBI.


Assuntos
Lesões Encefálicas/metabolismo , Córtex Cerebral/metabolismo , Microdomínios da Membrana/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Animais , Regulação da Expressão Gênica , Quinase I-kappa B , Masculino , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Receptor fas/metabolismo
7.
J Cereb Blood Flow Metab ; 25(11): 1505-16, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15959464

RESUMO

Recent evidence suggests that matrix metalloproteinases (MMPs) contribute to acute edema and lesion formation following ischemic and traumatic brain injuries (TBI). Experimental and clinical studies have also reported the beneficial effects of posttraumatic hypothermia on histopathological and behavioral outcome. The purpose of this study was to determine whether therapeutic hypothermia would affect the activity of MMPs after TBI. Male Sprague-Dawley rats were traumatized by moderate parasagittal fluid-percussion (F-P) brain injury. Seven groups (n=5/group) of animals were investigated: sham-operated, TBI with normothermia (37 degrees C), and TBI with hypothermia (33 degrees C). Normothermia animals were killed at 4, 24, 72 h and 5 days, and hypothermia animals at 24 or 72 h. Brain temperature was reduced to target temperature 30 mins after trauma and maintained for 4 h. Ipsilateral and contralateral cortical, hippocampal, and thalamic regions were analyzed by gelatin and in situ zymography. In traumatized normothermic animals, TBI significantly (P<0.005) increased MMP-9 levels in ipsilateral (right) cortical and hippocampal regions, compared with contralateral or sham animals, beginning at 4 h and persisting to 5 days. At 1, 3, and 5 days after TBI, significant increases in MMP-2 levels were observed. In contrast to these findings observed with normothermia, posttraumatic hypothermia significantly reduced MMP-9 levels. Hypothermic treatment, however, did not affect the delayed activation of MMP-2. Clarifying the mechanisms underlying the beneficial effects of posttraumatic hypothermia is an active area of research. Posttraumatic hypothermia may attenuate the deleterious consequences of brain trauma by reducing MMP activation acutely.


Assuntos
Lesões Encefálicas/enzimologia , Encéfalo/enzimologia , Hipotermia Induzida , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Animais , Encéfalo/patologia , Lesões Encefálicas/patologia , Lesões Encefálicas/terapia , Ativação Enzimática , Hipotermia Induzida/métodos , Masculino , Ratos , Ratos Sprague-Dawley
8.
Acta Otolaryngol ; 125(9): 922-8, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16109672

RESUMO

CONCLUSIONS: During systemic hypothermia, the internal temperature of the rat cochlea correlates best with the temporalis muscle and rectal temperatures. These positive correlations will be used in future studies to assess the efficacy of mild and moderate hypothermia to protect hearing against the progressive loss caused by electrode insertion in a clinically relevant model of cochlear implantation trauma. OBJECTIVE: To monitor the internal temperature of the cochlea during induced systemic hypothermia using a reference tissue instead of an internal cochlear temperature probe. MATERIAL AND METHODS: The temperatures of the cochlea, brain, temporalis muscle and rectum were determined during periods of normothermia (37 degrees C), mild (33 degrees C) and moderate (30 degrees C) hypothermia and slow rewarming in anesthetized adult Fisher rats. These values were compared using statistical analysis to establish the best correlations between the temperature of the cochlea and the temperature at the three other temperature measurement sites. RESULTS: The strongest correlations with the internal temperature of the cochlea during the induction of mild-to-moderate hypothermia were obtained for the temperatures of the ipsilateral temporalis muscle and rectum.


Assuntos
Temperatura Corporal , Cóclea/fisiologia , Perda Auditiva/prevenção & controle , Hipotermia Induzida , Reto/fisiologia , Músculo Temporal/fisiologia , Animais , Implantes Cocleares/efeitos adversos , Perda Auditiva/etiologia , Ratos , Ratos Endogâmicos F344 , Reaquecimento
9.
J Neurotrauma ; 32(4): 228-36, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25111533

RESUMO

The innate immune response contributes to the inflammatory activity after traumatic brain injury (TBI). In the present study we identify macrophage-inducible C-type lectin (mincle) as a pattern recognition receptor that contributes to innate immunity in neurons after TBI. Here we report that mincle is activated by SAP130 in cortical neurons in culture, resulting in production of the inflammatory cytokine TNF. In addition, mincle and SAP130 are elevated in the brain and cerebrospinal fluid of humans after TBI and the brain of rodents after fluid percussion brain injury. Thus, these findings suggest the involvement of mincle to the pathology of TBI. Importantly, blocking mincle with a neutralizing antibody against mincle in cortical neurons in culture treated with SAP130 resulted in inhibition of mincle signaling and decreased TNF production. Therefore, our findings identify mincle as a contributor to the inflammatory response after TBI.


Assuntos
Lesões Encefálicas/imunologia , Imunidade Inata/imunologia , Lectinas Tipo C/imunologia , Receptores Imunológicos/imunologia , Transdução de Sinais/imunologia , Adolescente , Adulto , Animais , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Ratos , Ratos Sprague-Dawley , Adulto Jovem
10.
J Inflamm (Lond) ; 12: 52, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26379474

RESUMO

BACKGROUND: Focal cerebral ischemia induces an inflammatory response that when exacerbated contributes to deleterious outcomes. The molecular basis regarding the regulation of the innate immune response after focal cerebral ischemia remains poorly understood. METHODS: In this study we examined the expression of retinoic acid-inducible gene (RIG)-like receptor-I (RIG-I) and its involvement in regulating inflammation after ischemia in the brain of rats subjected to middle cerebral artery occlusion (MCAO). In addition, we studied the regulation of RIG-I after oxygen glucose deprivation (OGD) in astrocytes in culture. RESULTS: In this study we show that in the hippocampus of rats, RIG-I and IFN-α are elevated after MCAO. Consistent with these results was an increased in RIG-I and IFN-α after OGD in astrocytes in culture. These data are consistent with immunohistochemical analysis of hippocampal sections, indicating that in GFAP-positive cells there was an increase in RIG-I after MCAO. In addition, in this study we have identified n-propyl gallate as an inhibitor of IFN-α signaling in astrocytes. CONCLUSION: Our findings suggest a role for RIG-I in contributing to the innate immune response after focal cerebral ischemia.

11.
J Cereb Blood Flow Metab ; 23(10): 1129-36, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14526223

RESUMO

XIAP is a member of the inhibitor of apoptosis (IAP) gene family that, in addition to suppressing cell death by inhibition and polyubiquitination of caspases, is involved in an increasing number of signaling cascades. Moreover, the function and regulation of XIAP in the central nervous system (CNS) is poorly understood. In this study, the authors investigated the cell-type expression, the subcellular distribution, ubiquitination of XIAP, and levels of Smac/DIABLO in the normal adult rat brain and in brains subjected to moderate traumatic brain injury (TBI). In the normal brain, XIAP was predominantly expressed in the perinuclear region of neurons. Traumatized brains showed dramatic alterations in cellular and regional expression of XIAP early after injury. Stereologic analyses of the number of XIAP-positive cells within the hippocampus of both hemispheres showed a biphasic response. Immunoprecipitation and immunoblots of extracts derived from different brain regions demonstrated that a single ubiquitin modifies XIAP. Normal cortex contained significantly higher levels of monoubiquitinated XIAP than hippocampus. TBI induced alterations in levels of monoubiquitinated XIAP that correlated with changes in XIAP distribution and immunoreactivity, suggesting that monoubiquitination of XIAP may be a regulator of XIAP location or activity. Similar levels of Smac/DIABLO were present in lysates of normal and traumatized brains. These data demonstrate for the first time a region-specific regulation of XIAP monoubiquitination in the normal adult rat brain, and after TBI, that may be a key event in the regulation of XIAP function contributing to the pathogenesis following injury.


Assuntos
Lesões Encefálicas/metabolismo , Neurônios/metabolismo , Proteínas/metabolismo , Ubiquitina/metabolismo , Animais , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose , Encéfalo/metabolismo , Lesões Encefálicas/patologia , Proteínas de Transporte/metabolismo , Imuno-Histoquímica , Masculino , Microscopia Confocal , Proteínas Mitocondriais/metabolismo , Neurônios/patologia , Biossíntese de Proteínas , Ratos , Ratos Sprague-Dawley , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X
12.
J Cereb Blood Flow Metab ; 24(8): 934-43, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15362724

RESUMO

Traumatic brain injury (TBI) leads to mossy fiber reorganization, which is considered to be a causative factor in the development of temporal lobe epilepsy. However, the underlying mechanism is not fully understood. Emerging evidence suggests that TrkB-ERK1/2-CREB/Elk-1 pathways are highly related to synaptic plasticity. This study used the rat fluid-percussion injury model to investigate activation of TrkB-ERK1/2-CREB/Elk-1 signaling pathways after TBI. Rats were subjected to 2.0-atm parasagittal TBI followed by 30 minutes, 4 hours, 24 hours, and 72 hours of recovery. After TBI, striking activation of TrkB-ERK1/2-CREB/Elk-1 signaling pathways in mossy fiber organization were observed with confocal microscopy and Western blot analysis. ERK1/2 was highly phosphorylated predominantly in hippocampal mossy fibers, whereas TrkB was phosphorylated both in the mossy fibers and the dentate gyrus region at 30 minutes and 4 hours of recovery after TBI. CREB was also activated at 30 minutes, peaked at 24 hours of recovery, and returned to the control level at 72 hours of recovery in dentate gyrus granule cells. Elk-1 phosphorylation was seen in CA3 neurons at 4 hours after TBI. The results suggest that the signaling pathways of TrkB-ERK1/2-CREB/Elk-1 are highly activated in mossy fiber organization, which may contribute to mossy fiber reorganization seen after TBI.


Assuntos
Lesões Encefálicas/enzimologia , Ativação Enzimática/fisiologia , Fibras Musgosas Hipocampais/enzimologia , Plasticidade Neuronal/fisiologia , Transdução de Sinais/fisiologia , Animais , Western Blotting , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Masculino , Microscopia Confocal , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fibras Musgosas Hipocampais/patologia , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor trkB/metabolismo , Fatores de Tempo , Fatores de Transcrição/metabolismo , Proteínas Elk-1 do Domínio ets
13.
Neurosurgery ; 51(1): 195-203; discussion 203, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12182417

RESUMO

OBJECTIVE: Posttraumatic temperature manipulations have been reported to significantly influence the inflammatory response to traumatic brain injury (TBI). The purpose of this study was to determine the temporal and regional profiles of messenger ribonucleic acid (mRNA) expression and protein levels for the proinflammatory cytokine interleukin-1beta (IL-1beta), after moderate or severe TBI. The effects of posttraumatic hypothermia (33 degrees C) or hyperthermia (39.5 degrees C) on these consequences of TBI were then determined. METHODS: Male Sprague-Dawley rats underwent fluid-percussion brain injury. In the first phase of the study, rats were killed 15 minutes or 1, 3, or 24 hours after moderate TBI (1.8-2.2 atmospheres), for reverse transcription-polymerase chain reaction analysis. Other groups of rats were killed 1, 3, 24, or 72 hours after moderate or severe TBI (2.4-2.7 atmospheres), for protein analysis. In the second phase, rats underwent moderate fluid-percussion brain injury, followed immediately by 3 hours of posttraumatic normothermia (37 degrees C), hyperthermia (39.5 degrees C), or hypothermia (33 degrees C), and were then killed, for analyses of protein levels and mRNA expression. Brain samples, including cerebral cortex, hippocampus, thalamus, and cerebellum, were dissected and stored at -80 degrees C until analyzed. RESULTS: The findings indicated that mRNA levels were increased (P < 0.05) as early as 1 hour after TBI and remained elevated up to 3 hours after moderate TBI. Although both moderate and severe TBI induced increased levels of IL-1beta (P < 0.05), increased protein levels were also noted in remote brain structures after severe TBI. Posttraumatic hypothermia attenuated IL-1beta protein levels, compared with normothermia (P < 0.05), although the levels remained elevated in comparison with sham values. In contrast, hyperthermia had no significant effect on IL-1beta levels, compared with normothermic values. Posttraumatic temperature manipulations had no significant effect on IL-1beta mRNA levels. CONCLUSION: Injury severity determines the degree of IL-1beta protein level elevation after TBI. The effects of posttraumatic hypothermia on IL-1beta protein levels (an important mediator of neurodegeneration after TBI) may partly explain the established effects of posttraumatic temperature manipulations on inflammatory processes after TBI.


Assuntos
Concussão Encefálica/genética , Interleucina-1/genética , RNA Mensageiro/genética , Animais , Encéfalo/imunologia , Encéfalo/patologia , Concussão Encefálica/imunologia , Concussão Encefálica/patologia , Córtex Cerebral/imunologia , Córtex Cerebral/patologia , Ensaio de Imunoadsorção Enzimática , Hipertermia Induzida , Hipotermia Induzida , Masculino , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
J Neurosurg ; 97(4): 945-53, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12405386

RESUMO

OBJECT: Although the benefits of posttraumatic hypothermia have been reported in experimental studies, the potential for therapeutic hypothermia to increase intracerebral hemorrhage remains a clinical concern. The purpose of this study was to quantify the amount of extravasated hemoglobin after traumatic brain injury (TBI) and to assess the changes in intracerebral hemoglobin concentrations under posttraumatic hypothermic and hyperthermic conditions. METHODS: Intubated and anesthetized rats were subjected to fluid-percussion injury (FPI). In the first experiment, rats were divided into moderate (1.8-2.2 atm) and severe (2.4-2.7 atm) TBI groups. In the second experiment, the effects of 3 hours of posttraumatic hypothermia (33 or 30 degrees C), hyperthermia (39 degrees C), or normothermia (37 degrees C) on hemoglobin levels following moderate trauma were assessed. The rats were perfused with saline at 24 hours postinjury, and then the traumatized and contralateral hemispheres, including the cerebellum, were dissected from whole brain. The hemoglobin level in each brain was quantified using a spectrophotometric hemoglobin assay. The results of these assays indicate that moderate and severe FPI induce increased levels of hemoglobin in the ipsilateral hemisphere (p < 0.0001). After severe TBI, the hemoglobin concentration was also significantly increased in the contralateral hemisphere (p < 0.05) and cerebellum (p < 0.005). Posttraumatic hypothermia (30 degrees C) attenuated hemoglobin levels (p < 0.005) in the ipsilateral hemisphere, whereas hyperthermia had a marked adverse effect on the hemoglobin concentration in the contralateral hemisphere (p < 0.05) and cerebellum (p < 0.005). CONCLUSIONS: Injury severity is an important determinant of the degree of hemoglobin extravasation after TBI. Posttraumatic hypothermia reduced hemoglobin extravasation, whereas hyperthermia increased hemoglobin levels compared with normothermia. These findings are consistent with previous data reporting that posttraumatic temperature manipulations alter the cerebrovascular and inflammatory consequences of TBI.


Assuntos
Temperatura Corporal , Lesões Encefálicas/metabolismo , Hemorragia Cerebral Traumática/metabolismo , Hemoglobinas/metabolismo , Animais , Lesões Encefálicas/terapia , Hemorragia Cerebral Traumática/terapia , Febre/metabolismo , Hipotermia Induzida , Masculino , Ratos , Ratos Sprague-Dawley , Índice de Gravidade de Doença , Espectrofotometria
15.
J Neurosurg ; 96(6): 1077-83, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12066910

RESUMO

OBJECT: Stilbazulenyl nitrone (STAZN) is a second-generation azulenyl nitrone that has markedly enhanced antioxidant properties compared with those of conventional alpha-phenyl nitrones. In this study, the authors assessed the potential efficacy of STAZN in a rodent model of fluid-percussion brain injury, which results in a consistent cortical contusion. METHODS: After anesthesia had been induced in normothermic Sprague-Dawley rats (brain temperature 36-36.5 degrees C) by halothane-nitrous oxide, the animals were subjected to a right parietooccipital parasagittal fluid-percussion injury (1.5-2 atm). The agent (STAZN, 30 mg/kg: eight animals) or vehicle (dimethyl sulfoxide; eight animals) was administered intraperitoneally at 5 minutes and 4 hours after trauma. The neurological status of each rat was evaluated on Days 1, 2, and 7 postinjury (normal score 0, maximum injury 12). Seven days after trauma, the rat brains were perfusion fixed, coronal sections at various levels were digitized, and areas of contusion were measured. Treatment with STAZN significantly improved neurological scores on Days 2 and 7 postinjury compared with vehicle-treated rats. Administration of STAZN also significantly reduced the total contusion area by 63% (1.8 +/- 0.5 mm2 in STAZN-treated animals compared with 4.8 +/- 2.1 mm2 in vehicle-treated animals; p = 0.04) and the deep cortical contusion area by 60% (1.2 +/- 0.2 mm2 in STAZN-treated animals compared with 2.9 +/- 1.2 mm2 in vehicle-treated animals; p = 0.03). By contrast, hippocampal cell loss in the CA3 sector was unaffected by STAZN treatment. CONCLUSIONS: Therapy with STAZN, a novel potent antioxidant, administered following traumatic brain injury, markedly improves neurological and histological outcomes. Azulenyl nitrones appear to represent a promising class of neuroprotective agents for combating this devastating condition.


Assuntos
Antioxidantes/uso terapêutico , Lesões Encefálicas/complicações , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/etiologia , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/etiologia , Fármacos Neuroprotetores/uso terapêutico , Óxidos de Nitrogênio/uso terapêutico , Sesquiterpenos/uso terapêutico , Animais , Azulenos , Lesões Encefálicas/patologia , Modelos Animais de Doenças , Masculino , Doenças do Sistema Nervoso/patologia , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
16.
J Neurotrauma ; 30(2): 119-30, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22963112

RESUMO

The midbrain median raphe (MR) and dorsal raphe (DR) nuclei were tested for their capacity to regulate recovery from traumatic brain injury (TBI). An implanted, wireless self-powered stimulator delivered intermittent 8-Hz pulse trains for 7 days to the rat's MR or DR, beginning 4-6 h after a moderate parasagittal (right) fluid-percussion injury. MR stimulation was also examined with a higher frequency (24 Hz) or a delayed start (7 days after injury). Controls had sham injuries, inactive stimulators, or both. The stimulation caused no apparent acute responses or adverse long-term changes. In water-maze trials conducted 5 weeks post-injury, early 8-Hz MR and DR stimulation restored the rate of acquisition of reference memory for a hidden platform of fixed location. Short-term spatial working memory, for a variably located hidden platform, was restored only by early 8-Hz MR stimulation. All stimulation protocols reversed injury-induced asymmetry of spontaneous forelimb reaching movements tested 6 weeks post-injury. Post-mortem histological measurement at 8 weeks post-injury revealed volume losses in parietal-occipital cortex and decussating white matter (corpus callosum plus external capsule), but not hippocampus. The cortical losses were significantly reversed by early 8-Hz MR and DR stimulation, the white matter losses by all forms of MR stimulation. The generally most effective protocol, 8-Hz MR stimulation, was tested 3 days post-injury for its acute effect on forebrain cyclic adenosine monophosphate (cAMP), a key trophic signaling molecule. This procedure reversed injury-induced declines of cAMP levels in both cortex and hippocampus. In conclusion, midbrain raphe nuclei can enduringly enhance recovery from early disseminated TBI, possibly in part through increased signaling by cAMP in efferent targets. A neurosurgical treatment for TBI using interim electrical stimulation in raphe repair centers is suggested.


Assuntos
Lesões Encefálicas/terapia , Terapia por Estimulação Elétrica/métodos , Núcleos da Rafe/fisiopatologia , Recuperação de Função Fisiológica , Animais , Comportamento Animal/fisiologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , AMP Cíclico/análise , AMP Cíclico/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Núcleos da Rafe/metabolismo , Núcleos da Rafe/patologia , Ratos , Ratos Sprague-Dawley
17.
Ther Hypothermia Temp Manag ; 2(4): 167-75, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23667780

RESUMO

Interleukin-6 (IL-6) is a proinflammatory cytokine that may play multiple roles in the pathogenesis of traumatic brain injury (TBI). The present study determined time-dependent changes in IL-6 concentrations in vulnerable brain regions, cerebrospinal fluid (CSF) samples, and plasma after normothermic TBI. Because secondary insults are common in head injured patients, we also assessed the consequences of a post-traumatic secondary hypoxic insult on this pleiotropic cytokine. Male Sprague-Dawley rats were intubated, anesthetized, and underwent a moderate parasagittal fluid-percussion brain injury (1.8-2.1 atm, 37°C) followed by either 30 minutes of normoxic or hypoxic (pO2 = 30-40 mmHg) gas levels. Rats were sacrificed 3, 6, or 24 hours after TBI or shamoperated procedures. Brain samples, including the ipsilateral cerebral cortex and hippocampus were dissected and analyzed. Plasma and CSF samples were collected at similar times and stored at -80°C until analysis. IL-6 levels were significantly increased ( p < 0.05) at 3, 6, and 24 hours in the cerebral cortex and at 6 hours in the hippocampus after TBI. IL-6 levels in the TBI normoxic group for both structures returned to control levels by 24 hours. Plasma levels of IL-6 were elevated at all time points, while CSF levels were high at 3 and 6 hours, but normalized by 24 hours. Post-traumatic hypoxia led to significantly elevated ( p < 0.05) IL-6 protein levels in the cerebral cortex at 24 hours compared to sham-operated controls. These findings demonstrate that moderate TBI leads to an early increase in IL-6 brain, plasma, and CSF protein levels. Secondary post-traumatic hypoxia, a common secondary injury mechanism, led to prolonged elevations in plasma IL-6 levels that may participate in the pathophysiology of this complicated TBI model.

18.
J Neurotrauma ; 29(2): 313-21, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22026555

RESUMO

The effects of slight variations in brain temperature on the pathophysiological consequences of acute brain injury have been extensively described in models of moderate and severe traumatic brain injury (TBI). In contrast, limited information is available regarding the potential consequences of temperature elevations on outcome following mild TBI (mTBI) or concussions. One potential confounding variable with mTBI is the presence of elevated body temperature that occurs in the civilian or military populations due to hot environments combined with exercise or other forms of physical exertion. We therefore determined the histopathological effects of pre- and post-traumatic hyperthermia (39°C) on mTBI. Adult male Sprague-Dawley rats were divided into 3 groups: pre/post-traumatic hyperthermia, post-traumatic hyperthermia alone for 2 h, and normothermia (37°C). The pre/post-hyperthermia group was treated with hyperthermia starting 15 min before mild parasagittal fluid-percussion brain injury (1.4-1.6 atm), with the temperature elevation extending for 2 h after trauma. At 72 h after mTBI, the rats were perfusion-fixed for quantitative histopathological evaluation. Contusion areas and volumes were significantly larger in the pre/post-hyperthermia treatment group compared to the post-hyperthermia and normothermic groups. In addition, pre/post-traumatic hyperthermia caused the most severe loss of NeuN-positive cells in the dentate hilus compared to normothermia. These neuropathological results demonstrate that relatively mild elevations in temperature associated with peri-traumatic events may affect the long-term functional consequences of mTBI. Because individuals exhibiting mildly elevated core temperatures may be predisposed to aggravated brain damage after mTBI or concussion, precautions should be introduced to target this important physiological variable.


Assuntos
Concussão Encefálica/patologia , Concussão Encefálica/fisiopatologia , Hipertermia Induzida/efeitos adversos , Neurônios/patologia , Animais , Concussão Encefálica/complicações , Lesões Encefálicas/complicações , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Modelos Animais de Doenças , Febre/etiologia , Febre/patologia , Febre/fisiopatologia , Hipertermia Induzida/métodos , Masculino , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley , Tempo de Reação/fisiologia , Fatores de Tempo
19.
Exp Neurol ; 233(2): 821-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22197046

RESUMO

Previous studies have demonstrated that moderate hypothermia reduces histopathological damage and improves behavioral outcome after experimental traumatic brain injury (TBI). Further investigations have clarified the mechanisms underlying the beneficial effects of hypothermia by showing that cooling reduces multiple cell injury cascades. The purpose of this study was to determine whether hypothermia could also enhance endogenous reparative processes following TBI such as neurogenesis and the replacement of lost neurons. Male Sprague-Dawley rats underwent moderate fluid-percussion brain injury and then were randomized into normothermia (37°C) or hypothermia (33°C) treatment. Animals received injections of 5-bromo-2'-deoxyuridine (BrdU) to detect mitotic cells after brain injury. After 3 or 7 days, animals were perfusion-fixed and processed for immunocytochemistry and confocal analysis. Sections were stained for markers selective for cell proliferation (BrdU), neuroblasts and immature neurons (doublecortin), and mature neurons (NeuN) and then analyzed using non-biased stereology to quantify neurogenesis in the dentate gyrus (DG). At 7 days after TBI, both normothermic and hypothermic TBI animals demonstrated a significant increase in the number of BrdU-immunoreactive cells in the DG as compared to sham-operated controls. At 7 days post-injury, hypothermia animals had a greater number of BrdU (ipsilateral cortex) and doublecortin (ipsilateral and contralateral cortex) immunoreactive cells in the DG as compared to normothermia animals. Because adult neurogenesis following injury may be associated with enhanced functional recovery, these data demonstrate that therapeutic hypothermia sustains the increase in neurogenesis induced by TBI and this may be one of the mechanisms by which hypothermia promotes reparative strategies in the injured nervous system.


Assuntos
Lesões Encefálicas/metabolismo , Giro Denteado/metabolismo , Hipotermia Induzida , Proteínas Associadas aos Microtúbulos/biossíntese , Neurogênese/fisiologia , Neurônios/metabolismo , Neuropeptídeos/biossíntese , Animais , Lesões Encefálicas/patologia , Lesões Encefálicas/terapia , Giro Denteado/citologia , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Regulação da Expressão Gênica , Hipotermia Induzida/métodos , Imuno-Histoquímica , Masculino , Proteínas Associadas aos Microtúbulos/genética , Neuropeptídeos/genética , Ratos , Ratos Sprague-Dawley
20.
Brain Res ; 1431: 23-34, 2012 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-22137658

RESUMO

Postoperative cognitive dysfunction, POCD, afflicts a large number of elderly surgical patients following surgery with general anesthesia. Mechanisms of POCD remain unclear. N-methyl-D-aspartate (NMDA) receptors, critical in learning and memory, that display protein expression changes with age are modulated by inhalation anesthetics. The aim of this study was to identify protein expression changes in NMDA receptor subunits and downstream signaling pathways in aged rats that demonstrated anesthesia-induced spatial learning impairments. Three-month-old and 18-month-old male Fischer 344 rats were randomly assigned to receive 1.8% isoflurane/70% nitrous oxide (N(2)O) anesthesia for 4h or no anesthesia. Spatial learning was assessed at 2weeks and 3months post-anesthesia in Morris water maze. Hippocampal and cortical protein lysates of 18-month-old rats were immunoblotted for activated caspase 3, NMDA receptor subunits, and extracellular-signal regulated kinase (ERK) 1/2. In a separate experiment, Ro 25-6981 (0.5mg/kg dose) was administered by I.P. injection before anesthesia to 18-month-old rats. Immunoblotting of NR2B was performed on hippocampal protein lysates. At 3months post-anesthesia, rats treated with anesthesia at 18-months-old demonstrated spatial learning impairment corresponding to acute and long-term increases in NR2B protein expression and a reduction in phospho-ERK1/2 in the hippocampus and cortex. Ro 25-6981 pretreatment attenuated the increase in acute NR2B protein expression. Our findings suggest a role for disruption of NMDA receptor mediated signaling pathways in the hippocampus and cortex of rats treated with isoflurane/ N(2)O anesthesia at 18-months-old, leading to spatial learning deficits in these animals. A potential therapeutic intervention for anesthesia associated cognitive deficits is discussed.


Assuntos
Envelhecimento/efeitos dos fármacos , Anestésicos Inalatórios/metabolismo , Encéfalo/efeitos dos fármacos , Isoflurano/farmacologia , Óxido Nitroso/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Envelhecimento/metabolismo , Análise de Variância , Animais , Encéfalo/citologia , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Interações Medicamentosas , Antagonistas de Aminoácidos Excitatórios/farmacologia , Masculino , Neurônios/metabolismo , Fenóis/farmacologia , Piperidinas/farmacologia , Ratos , Ratos Endogâmicos F344 , Tempo de Reação/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/genética , Comportamento Espacial/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA