Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Acta Neurochir (Wien) ; 148(2): 181-93; discussion 193-4, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16362181

RESUMO

Traumatic brain injury (TBI) evokes widespread/diffuse axonal injury (TAI) significantly contributing to its morbidity and mortality. While classic theories suggest that traumatically injured axons are mechanically torn at the moment of injury, studies in the last two decades have not supported this premise in the majority of injured axons. Rather, current thought considers TAI a progressive process evoked by the tensile forces of injury, gradually evolving from focal axonal alteration to ultimate disconnection. Recent observations have demonstrated that traumatically induced focal axolemmal permeability leads to local influx of Ca2+ with the subsequent activation of the cysteine proteases, calpain and caspase, that then play a pivotal role in the ensuing pathogenesis of TAI via proteolytic digestion of brain spectrin, a major constituent of the subaxolemmal cytoskeletal network, the "membrane skeleton". In this pathological progression this local Ca2+ overloading with the activation of calpains also initiates mitochondrial injury that results in the release of cytochrome-c, with the activation of caspase. Both the activated calpain and caspases then participate in the degradation of the local axonal cytoskeleton causing local axonal failure and disconnection. In this review, we summarize contemporary thought on the pathogenesis of TAI, while discussing the potential diversity of pathological processes observed within various injured fiber types. The anterograde and retrograde consequences of TAI are also considered together with a discussion of various experimental therapeutic approaches capable of attenuating TAI.


Assuntos
Axônios/patologia , Lesões Encefálicas/fisiopatologia , Encéfalo/fisiopatologia , Lesão Axonal Difusa/fisiopatologia , Degeneração Walleriana/fisiopatologia , Axônios/metabolismo , Encéfalo/patologia , Cálcio/metabolismo , Calpaína/metabolismo , Caspases/metabolismo , Morte Celular/fisiologia , Citoesqueleto/metabolismo , Citoesqueleto/patologia , Humanos
2.
Acta Neurochir (Wien) ; 147(8): 855-61, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15924207

RESUMO

BACKGROUND: Calcium-induced proteolytic processes are considered key players in the progressive pathobiology of traumatic brain injury (TBI). Activation of calpain and caspases after TBI leads to the cleavage of cytoskeletal proteins such as non-erythroid alpha II-spectrin. Recent reports demonstrate that the levels of spectrin and spectrin breakdown products (SBDPs) are elevated in vitro after mechanical injury, in the cerebrospinal fluid (CSF) and brain tissue following experimental TBI, and in human brain tissue after TBI. METHODS: This study was initiated to detect spectrin and SBDP accumulation in the ventricular CSF of 12 severe TBI-patients with raised intracranial pressure (ICP). Nine patients with non-traumatically elevated ICP and 5 undergoing diagnostic lumbar puncture (LP) served as controls. Intact spectrin and calpain and caspase specific SBDPs in CSF collected once a day over a several day period were assessed via Western blot analysis. Parameters of severity and outcome such as ICP, Glasgow Coma Scale and Glasgow Outcome Scale were also monitored in order to reveal a potential correlation between these CSF markers and clinical parameters. RESULTS: In control patients undergone LP no immunoreactivity was detected. Non-erythroid alpha-II-spectrin and SBDP occurred more frequently and their level was significantly higher in the CSF of TBI patients than in other pathological conditions associated with raised ICP. Those TBI patients followed for several days post-injury revealed a consistent temporal pattern for protein accumulation with the highest level achieved on the 2(nd) -3(rd) days after TBI. CONCLUSION: Elevation of calpain and caspase specific SBDPs is a significant finding in TBI patients indicating that intact brain spectrin- and SBDP-levels are closely associated with the specific neurochemical processes evoked by TBI. The results strongly support the potential utility of these surrogate markers in the clinical monitoring of patients with severe TBI and provide further evidence of the role of calcium-induced, calpain- and caspase-mediated structural proteolysis in TBI.


Assuntos
Lesões Encefálicas/líquido cefalorraquidiano , Pressão Intracraniana/fisiologia , Espectrina/líquido cefalorraquidiano , Adolescente , Adulto , Idoso , Estudos de Casos e Controles , Ventrículos Cerebrais/metabolismo , Feminino , Escala de Coma de Glasgow , Escala de Resultado de Glasgow , Humanos , Masculino , Pessoa de Meia-Idade , Espectrina/metabolismo , Fatores de Tempo
3.
J Neurotrauma ; 20(3): 261-8, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12820680

RESUMO

Traumatic brain injury (TBI) evokes diffuse (traumatic) axonal injury (TAI), which contributes to morbidity and mortality. Damaged axons display progressive alterations gradually evolving to axonal disconnection. In severe TAI, the tensile forces of injury lead to a focal influx of Ca2+, initiating a series of proteolytic processes wherein the cysteine proteases, calpain and caspase modify the axonal cytoskeleton, causing irreversible damage over time postinjury. Although several studies have demonstrated that the systemic administration of calpain inhibitors reduces the extent of ischemic and traumatic contusional injury a direct beneficial effect on TAI has not been established to date. The current study was initiated to address this issue in an impact acceleration rat-TBI model in order to provide further evidence on the contribution of calpain-mediated proteolytic processes in the pathogenesis of TAI, while further supporting the utility of calpain-inhibitors. A single tail vein bolus injection of 30 mg/kg MDL-28170 was administered to Wistar rats 30 min preinjury. After injury the rats were allowed to survive 120 min when they were perfused with aldehydes. Brains were processed for immunohistochemical localization of damaged axonal profiles displaying either amyloid precursor protein (APP)- or RMO-14-immunoreactivity (IR), both considered markers of specific features of TAI. Digital data acquisition and statistical analysis demonstrated that preinjury administration of MDL-28170 significantly reduced the mean number of damaged RMO-14- as well as APP-IR axonal profiles in the brainstem fiber tracts analyzed. These results further underscore the role of calpain-mediated proteolytic processes in the pathogenesis of DAI and support the potential use of cell permeable calpain-inhibitors as a rational therapeutic approach in TBI.


Assuntos
Axônios/efeitos dos fármacos , Axônios/patologia , Lesões Encefálicas/tratamento farmacológico , Inibidores de Cisteína Proteinase/farmacologia , Dipeptídeos/farmacologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Axônios/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Modelos Animais , Proteínas de Neurofilamentos/metabolismo , Ratos , Ratos Wistar
4.
J Neurotrauma ; 18(10): 993-1009, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11686499

RESUMO

Traumatic brain injury (TBI) is documented to have detrimental effects on CNS metabolism, including alterations in glucose utilization and the depression of mitochondrial oxidative phosphorylation. Studies on mitochondrial metabolism have also provided evidence for reduced activity of the cytochrome oxidase complex of the electron transport chain (complex IV) after TBI and an immediate (lhr) reduction in mitochondrial state 3 respiratory rate, which can persist for up to 14 days postinjury. Using differential display methods to screen for differences in gene expression, we have found that cytochrome c oxidase II (COII), a mitochondrial encoded subunit of complex IV, is upregulated following TBI. Since COII carries a binding site for cytochrome c in the respiratory chain, and since it is required for the passage of chain electrons to molecular oxygen, driving the production of ATP, we hypothesized that metabolic dysfunction resulting from TBI alters COII gene expression directly, perhaps influencing the synaptic plasticity that occurs during postinjury recovery processes. To test this hypothesis, we documented COII mRNA expression and complex IV (cytochrome c oxidase) functional activity at 7 days postinjury, focusing on the long-term postinjury period most closely associated with synaptic reorganization. Both central fluid percussion TBI and combined TBI and bilateral entorhinal cortical lesion were examined. At 7 days survival, differential display, RT-PCR, and Northern blot analysis of hippocampal RNA from both TBI and combined insult models showed a significant induction of COII mRNA. This long-term elevation in COII gene expression was supported by increases in COII immunobinding. By contrast, cytochrome oxidase histochemical activity within tissue sections from injured brains suggested a reduction of complex IV activity within the TBI cases, but not within animals subjected to the combined insult. These differences in cytochrome c oxidase activity were supported by in vitro assay of complex IV using cerebral cortical and hippocampal tissues. Our present results support the hypothesis that COII is selectively vulnerable to TBI and that COII differences may indicate the degree of metabolic dysfunction induced by different pathologies. Taken together, such data will better define the role of metabolic function in long-term recovery after TBI.


Assuntos
Lesões Encefálicas/genética , Lesões Encefálicas/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Regulação Enzimológica da Expressão Gênica/genética , Mitocôndrias/enzimologia , Animais , Northern Blotting , Clonagem Molecular , Metabolismo Energético/fisiologia , Córtex Entorrinal/patologia , Imuno-Histoquímica , Masculino , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/isolamento & purificação , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Exp Neurol ; 172(2): 320-31, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11716556

RESUMO

Traumatic axonal injury (TAI) contributes to morbidity and mortality following traumatic brain injury (TBI). Single-label immunocytochemical studies employing antibodies to neurofilament compaction (NFC), RM014, and antibodies to APP, a marker of impaired axonal transport (AxT), have shown that TAI involves both NFC and disruption of AxT. Although it may be hypothesized that both events occur within the same injured axon, this has not been confirmed. To determine the relationship between NFC and impaired AxT, dual-label immunofluorescence was employed. To compare and contrast specific changes associated with these two markers of TAI, single-label electron microscopy was also used. Rats were subjected to an impact acceleration injury (30 min-6 h survival), and their brains were prepared for dual-label immunofluorescence and single-label electron microscopy. APP and RM014 were consistently found in two distinct classes of TAI. One, which showed only RM014 immunoreactivity, was thin and elongate, was sometimes vacuolated, and revealed little progressive change over time. The second was distinguished by focal axonal swellings containing APP immunoreactivity alone in small-caliber axons or in combination with RM014 immunoreactivity in large-caliber axons. These swellings were localized to either nodal or internodal loci and underwent progressive swelling over time, ultimately leading to secondary axotomy. Ultrastructural examination of these two classes of TAI revealed NFC together with mitochondrial dilation without organelle pooling in the RM014 single-labeled axons. However, the APP single-labeled small-caliber axons and APP/RM014 dual-labeled large-caliber axons revealed a progressive accumulation of organelles associated with increased axonal swelling over time. In contrast to previous thought, it now appears that NFC may occur independent of impaired AxT in TAI. This finding underscores the complexity of TAI, suggesting the need for multiple immunocytochemical approaches to fully assess the overall axonal response to TBI.


Assuntos
Axônios/patologia , Edema Encefálico/etiologia , Edema Encefálico/patologia , Lesões Encefálicas/complicações , Lesão Axonal Difusa/complicações , Fibras Nervosas/patologia , Ferimentos não Penetrantes/complicações , Animais , Imuno-Histoquímica , Masculino , Microscopia Eletrônica , Tratos Piramidais/patologia , Ratos , Ratos Sprague-Dawley , Valores de Referência
6.
Exp Neurol ; 172(1): 199-210, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11681852

RESUMO

Our laboratory has shown that traumatically induced axonal injury (TAI) is significantly reduced by posttraumatic hypothermia followed by slow rewarming. Further, TAI can be exacerbated by rapid rewarming, and the damaging consequences of rapid rewarming can be reversed by cyclosporin A, which is believed to protect via blunting mitochondrial permeability transition (MPT). In this communication, we continue investigating the damaging consequences of rapid posthypothermic rewarming and the protective role of immunophilin ligands using another member of the immunophilin family, FK506, which does not affect MPT but rather inhibits calcineurin. Rats were subjected to impact-acceleration brain injury followed by the induction of hypothermia with subsequent rapid or slow posthypothermic rewarming. During rewarming, animals received either FK506 or its vehicle. Three hours postinjury, animals were prepared for the visualization of TAI via antibodies targeting impaired axoplasmic transport (APP) and/or overt neurofilament alteration (RMO-14). Rapid rewarming exacerbated TAI, which was attenuated by FK506. This protection was statistically significant for the APP-immunoreactive fibers but not for the RMO-14-positive fibers. Combined labeling, using one chromagen to visualize both axonal changes, suggested that these two immunoreactive profiles revealed two distinct pathologies not occurring along the same axon. Collectively, these studies confirmed previous observations identifying the adverse consequences of rapid rewarming while also showing the complexity of the pathobiology of TAI. Additionally, the demonstration that FK506 is protective suggests that calcineurin may be a major target for neuroprotection.


Assuntos
Axônios/efeitos dos fármacos , Lesões Encefálicas/prevenção & controle , Hipotermia Induzida , Imunofilinas , Reaquecimento/efeitos adversos , Tacrolimo/farmacologia , Animais , Transporte Axonal/efeitos dos fármacos , Axônios/metabolismo , Axônios/patologia , Pressão Sanguínea , Temperatura Corporal , Lesões Encefálicas/patologia , Inibidores de Calcineurina , Contagem de Células , Imuno-Histoquímica , Imunofilinas/antagonistas & inibidores , Imunossupressores/farmacologia , Ligantes , Masculino , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley , Reaquecimento/métodos , Fatores de Tempo
7.
J Neurotrauma ; 18(6): 607-14, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11437083

RESUMO

The immunophilin ligand, cyclosporin A (CsA), is effective in reducing the axonal damage associated with traumatic brain injury (TBI). Based upon extensive ultrastructural and immunohistochemical studies, the neuroprotection afforded by CsA appeared to be mediated via mitochondrial protection, specifically, the prevention of mitochondrial swelling and inhibition of mitochondrial permeability transition (MPT). However, the potential that CsA could also be neuroprotective via the immunophilin-mediated inhibition of the protein phosphatase, calcineurin (CN) has not been directly assessed. To address this issue, the current study assessed the ability of FK506, another immunophilin ligand that inhibits CN with no effect on MPT, to attenuate axonal damage in a rat impact-acceleration model of TBI. Traumatic axonal injury (TAI), detected via an antibody against beta-amyloid precursor protein (APP), a specific marker of axonal injury, was significantly reduced at 24 hr postinjury in Sprague-Dawley rats receiving intravenous FK506 (2 mg/kg; n = 5) 30 min prior to injury compared to vehicle controls (n = 3). While not rejecting the established efficacy of CsA in providing neuroprotection via its targeting of MPT, this study does underscore the potential importance of CN in the progressive pathobiology of TAI, suggesting that CN may constitute another important therapeutic target.


Assuntos
Axônios/patologia , Lesões Encefálicas/tratamento farmacológico , Imunossupressores/uso terapêutico , Tacrolimo/uso terapêutico , Aceleração , Animais , Gasometria , Encéfalo/metabolismo , Lesões Encefálicas/patologia , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Imunossupressores/farmacocinética , Injeções Intravenosas , Masculino , Ratos , Ratos Sprague-Dawley , Tacrolimo/farmacocinética
8.
J Neurosurg ; 94(3): 493-8, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11235956

RESUMO

OBJECT: Although considerable attention has been focused on the use of posttraumatic hypothermia, little consideration has been given to the issue of posthypothermic rewarming and its potentially damaging consequences. In this communication, the authors examine the issue of rapid posthypothermic rewarming compared with gradual rewarming while exploring the potential utility of cyclosporin A (CsA) administration for attenuating any rapid rewarming-induced axonal change. METHODS: Male Sprague-Dawley rats were subjected to impact-acceleration injury and then their body temperature was lowered to 32 degrees C for 1 hour postinjury. After hypothermia, rewarming to normothermic levels was accomplished either within a 20-minute period (rapid rewarming) or over a 90-minute period (slow rewarming). Some animals in the rapid rewarming group received intrathecal infusion of either CsA or its vehicle, whereas the rats in the slow rewarming group received vehicle alone. Both the CsA and its vehicle were administered immediately before initiation of rewarming. Twenty-four hours postinjury the animals' brains were processed for visualization of amyloid precursor protein (APP), a marker of traumatic axonal injury. The APP-positive axonal density in the gradually rewarmed group receiving vehicle was statistically significantly reduced in comparison with the rapidly rewarmed, vehicle-treated group. For the group undergoing rapid rewarming and treatment with CsA, a statistically significant reduction was also found in the density of the APP profiles compared with the rapidly rewarmed, vehicle-treated group. CONCLUSIONS: The results of this study show that rapid rewarming exacerbates traumatically induced axonal injury, which can be significantly attenuated by administering CsA.


Assuntos
Axônios/patologia , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/patologia , Ciclosporina/farmacologia , Hipotermia Induzida/efeitos adversos , Imunossupressores/farmacologia , Precursor de Proteína beta-Amiloide/análise , Animais , Axônios/química , Axônios/efeitos dos fármacos , Temperatura Corporal , Hipotermia Induzida/métodos , Imuno-Histoquímica , Masculino , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
9.
J Neurotrauma ; 18(1): 47-55, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11200249

RESUMO

This study examined whether NMDA-stimulated cyclic GMP levels were altered at two different time points following lateral fluid percussion injury. At 60 min and 15 days postinjury, the left and right hippocampi were dissected and chopped into mini-prisms. Each hippocampus was divided into five equal parts and incubated with either the phosphodiesterase inhibitor IBMX (3-isobutyl-1-methylxanthine, 500 microM) alone, IBMX and N-methyl-D-aspartic acid (NMDA) OR IBMX, NMDA, and glycine (10 MM). Two concentrations of NMDA were used: 500 or 1,000 microM. Tissues were then assayed for levels of cyclic GMP. Results indicated that there were no changes in basal levels of cyclic GMP at either postinjury time point. At 60 min postinjury, there were no significant main effects for injury or drug concentration. There was a significant injury x side interaction effect with increased levels of NMDA-stimulated cyclic GMP in the hippocampus ipsilateral to the injury impact and decreased cyclic GMP levels in the contralateral hippocampus. There were no significant alterations in NMDA-stimulated cyclic GMP levels at 15 days postinjury. The data from this study indicated that NMDA-stimulated cyclic GMP accumulation is differentially altered in the hippocampus ipsilateral and contralateral to the site of the injury at 1 h after injury, but is normalized by 15 days postinjury. These findings implicate NMDA-mediated intracellular signaling processes in the acute excitotoxic response to injury.


Assuntos
Lesões Encefálicas/metabolismo , GMP Cíclico/metabolismo , Lateralidade Funcional/fisiologia , Hipocampo/metabolismo , N-Metilaspartato/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Masculino , Percussão/efeitos adversos , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Fatores de Tempo
10.
Exp Neurol ; 166(1): 136-52, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11031090

RESUMO

The rat model of combined central fluid percussion traumatic brain injury (TBI) and bilateral entorhinal cortical lesion (BEC) produces profound, persistent cognitive deficits, sequelae associated with human TBI. In contrast to percussive TBI alone, this combined injury induces maladaptive hippocampal plasticity. Recent reports suggest a potential role for dopamine in CNS plasticity after trauma. We have examined the effect of the dopamine enhancer l-deprenyl on cognitive function and neuroplasticity following TBI. Rats received fluid percussion TBI, BEC alone, or combined TBI + BEC lesion and were treated once daily for 7 days with l-deprenyl, beginning 24 h after TBI alone and 15 min after BEC or TBI + BEC. Postinjury motor assessment showed no effect of l-deprenyl treatment. Cognitive performance was assessed on days 11-15 postinjury and brains from the same cases examined for dopamine beta-hydroxylase immunoreactivity (DBH-IR) and acetylcholinesterase (AChE) histochemistry. Significant cognitive improvement relative to untreated injured cases was observed in both TBI groups following l-deprenyl treatment; however, no drug effects were seen with BEC alone. l-Deprenyl attenuated injury-induced loss in DBH-IR over CA1 and CA3 after TBI alone. However, after combined TBI + BEC, l-deprenyl was only effective in protecting CA1 DBH-IR. AChE histostaining in CA3 was significantly elevated with l-deprenyl in both injury models. After TBI + BEC, l-deprenyl also increased AChE in the dentate molecular layer relative to untreated injured cases. These results suggest that dopaminergic/noradrenergic enhancement facilitates cognitive recovery after brain injury and that noradrenergic fiber integrity is correlated with enhanced synaptic plasticity in the injured hippocampus.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Transtornos Cognitivos/tratamento farmacológico , Plasticidade Neuronal/efeitos dos fármacos , Recuperação de Função Fisiológica/efeitos dos fármacos , Selegilina/farmacologia , Acetilcolinesterase/metabolismo , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Axônios/patologia , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Cognição/efeitos dos fármacos , Cognição/fisiologia , Transtornos Cognitivos/patologia , Transtornos Cognitivos/fisiopatologia , Modelos Animais de Doenças , Dopamina beta-Hidroxilase/metabolismo , Córtex Entorrinal/lesões , Córtex Entorrinal/patologia , Córtex Entorrinal/fisiopatologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipocampo/fisiopatologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Vias Neurais/lesões , Vias Neurais/patologia , Vias Neurais/fisiopatologia , Plasticidade Neuronal/fisiologia , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/fisiologia , Fatores de Tempo
12.
Brain Res ; 871(2): 288-302, 2000 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-10899295

RESUMO

Antibodies to the amyloid precursor protein (APP) are commonly used to detect traumatic axonal injury (TAI). Carried by fast anterograde axoplasmic transport, APP will pool at regions of impaired transport associated with TAI. Based primarily upon commercial antibody availability, previous studies have targeted the N-terminus of APP, which, with respect to antigen detection, is suboptimally located within anterogradely transported vesicles. Recently, antibodies to the APP C-terminus, located on the external surface of anterogradely transported vesicles, have become available, allowing for the exploration of their utility in detecting TAI. To this end, rats were subjected to an impact acceleration injury, surviving 30 min to 24 h post-injury. They were then perfused, their brains sectioned and prepared for dual label immunofluorescent microscopy, single label bright field microscopy, and electron microscopy (EM). Antibodies to the APP C-terminus yielded the ready detection of intensely labeled TAI with significantly reduced diffuse background staining in comparison to antibodies to the APP N-terminus in both dual label immunofluorescent and single label bright-field approaches. EM examination of antibodies to the APP C-terminus in TAI revealed intense labeling of pooled intra-axonal vesicular profiles, confirming the anterogradely transported vesicular source of the APP seen in TAI. Interestingly, in addition to providing a technically superior approach and new detailed information on the subcellular localization of APP, antibodies to the APP C-terminus also proved more cost effective. Immunofluorescent studies of APP C-terminus immunoreactivity involved 1/3 the cost of targeting the N-terminus, while bright field APP C-terminus studies were performed for 1/20 the cost.


Assuntos
Precursor de Proteína beta-Amiloide/imunologia , Anticorpos , Axônios/imunologia , Lesões Encefálicas/imunologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Axônios/patologia , Axônios/ultraestrutura , Biomarcadores , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Imunofluorescência , Masculino , Microscopia Eletrônica , Estrutura Terciária de Proteína , Ratos
13.
J Neurosci Res ; 60(3): 370-9, 2000 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10797540

RESUMO

Pathological processes affecting presynaptic terminals may contribute to morbidity following traumatic brain injury (TBI). Posttraumatic widespread neuronal depolarization and elevated extracellular potassium and glutamate are predicted to alter the transduction of action potentials in terminals into reliable synaptic transmission and postsynaptic excitation. Evoked responses to orthodromic single- and paired-pulse stimulation were examined in the CA1 dendritic region of hippocampal slices removed from adult rats following fluid percussion TBI. The mean duration of the extracellularly recorded presynaptic volley (PV) increased from 1.08 msec in controls to 1.54 msec in slices prepared at 1 hr postinjury. There was a time-dependent recovery of this injury effect, and PV durations at 2 and 7 days postinjury were not different from controls. In slices removed at 1 hr postinjury, the initial slopes of field excitatory postsynaptic potentials (fEPSPs) were reduced to 36% of control values, and input/output plots revealed posttraumatic deficits in the transfer of excitation from pre- to postsynaptic elements. Manipulating potassium currents with 1.0 mM tetraethylammonium or elevating potassium ion concentration to 7.5 mM altered evoked responses but did not replicate the injury effects to PV duration. Paired-pulse facilitation of fEPSP slopes was significantly elevated at all postinjury survivals: 1 hr, 2 days, and 7 days. These results suggest two pathological processes with differing time courses: 1) a transient impairment of presynaptic terminal functioning affecting PV durations and the transduction of afferent activity in the terminals to reliable synaptic excitation and 2) a more protracted deficit to the plasticity mechanisms underlying paired-pulse facilitation.


Assuntos
Lesões Encefálicas/fisiopatologia , Receptores Pré-Sinápticos/fisiologia , Animais , Cálcio/metabolismo , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/metabolismo , Estimulação Elétrica , Eletrodos Implantados , Eletrofisiologia , Potenciais Evocados/fisiologia , Ativação do Canal Iônico/fisiologia , Masculino , Plasticidade Neuronal/fisiologia , Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Pré-Sinápticos/metabolismo , Sistemas do Segundo Mensageiro/efeitos dos fármacos
14.
J Histochem Cytochem ; 48(1): 153-61, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10653596

RESUMO

Fluorescent immunocytochemistry (FICC) allows multiple labeling approaches when enzyme-based techniques are difficult to combine, such as in double-labeling experiments targeting small-caliber axonal segments. Nevertheless, the conversion of FICC to a product visible at the electron microscopic (EM) level requires labor-intensive procedures, thus justifying the development of more user-friendly conversion methods. This study was initiated to simplify the conversion of FICC to EM by employing the unique properties of tyramide signal amplification (TSA), which allowed the simultaneous targeting of a fluorescent tag and biotin label to the same antigenic site. Briefly, one of two antigenic sites typically co-localized in damaged axonal segments was visualized by the application of a fluorescent secondary antibody, with the other tagged via a biotinylated antibody. Next, an ABC kit was used, followed by the simultaneous application of fluorophore-tyramide and biotin-tyramide. After temporary mounting for fluorescent digital photomicroscopy, sections were incubated in ABC and reacted with diaminobenzidine before EM analysis. Double-labeling fluorescent immunocytochemistry with TSA clearly delineated damaged axonal segments. In addition, these same axonal segments yielded high-quality EM images with discrete electron-dense reaction products, thereby providing a simple and reproducible means for following fluorescent analysis with EM.


Assuntos
Axônios/ultraestrutura , Imunofluorescência , Corantes Fluorescentes , Microscopia Imunoeletrônica/métodos , Tiramina/análogos & derivados , Animais , Axônios/patologia , Biotina/análogos & derivados , Traumatismos Craniocerebrais/patologia , Processamento de Imagem Assistida por Computador , Ratos , Ratos Sprague-Dawley , Rodaminas , Traumatismos do Sistema Nervoso/patologia
15.
J Neurosci ; 20(8): 2825-34, 2000 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10751434

RESUMO

Axonal injury is a feature of traumatic brain injury (TBI) contributing to both morbidity and mortality. The traumatic axon injury (TAI) results from focal perturbations of the axolemma, allowing for calcium influx triggering local intraaxonal cytoskeletal and mitochondrial damage. This mitochondrial damage has been posited to cause local bioenergetic failure, leading to axonal failure and disconnection; however, this mitochondrial damage may also lead to the release of cytochrome c (cyto-c), which then activates caspases with significant adverse intraaxonal consequences. In the current communication, we examine this possibility. Rats were subjected to TBI, perfused with aldehydes at 15-360 min after injury, and processed for light microscopic (LM) and electron microscopic (EM) single-labeling immunohistochemistry to detect extramitochondrially localized cytochrome c (cyto-c) and the signature protein of caspase-3 activation (120 kDa breakdown product of alpha-spectrin) in TAI. Combinations of double-labeling fluorescent immunohistochemistry (D-FIHC) were also used to demonstrate colocalization of calpain activation with cyto-c release and caspase-3-induction. In foci of TAI qualitative-quantitative LM demonstrated a parallel, significant increase in cyto-c release and caspase-3 activation over time after injury. EM analysis demonstrated that cyto-c and caspase-3 immunoreactivity were associated with mitochondrial swelling-disruption in sites of TAI. Furthermore, D-IFHC revealed a colocalization of calpain activation, cyto-c release, and caspase-3 induction in these foci, which also revealed progressive TAI. The results demonstrate that cyto-c and caspase-3 participate in the terminal processes of TAI. This suggests that those factors that play a role in the apoptosis in the neuronal soma are also major contributors to the demise of the axonal appendage.


Assuntos
Lesões Encefálicas/enzimologia , Calpaína/metabolismo , Caspases/metabolismo , Grupo dos Citocromos c/metabolismo , Lesão Axonal Difusa/enzimologia , Animais , Lesões Encefálicas/patologia , Caspase 3 , Lesão Axonal Difusa/patologia , Ativação Enzimática , Ratos , Ratos Sprague-Dawley
16.
Exp Neurol ; 159(1): 319-28, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10486200

RESUMO

Traumatic brain injury (TBI) in animals and man generates widespread axonal injury characterized by focal axolemmal permeability changes, induction of calpain-mediated proteolysis, and neurofilament side-arm modification associated with neurofilament compaction (NFC) evolving to axonal disconnection. Recent observations have suggested that moderate hypothermia is neuroprotective in several models of TBI. Nevertheless, the pathway by which hypothermia prevents traumatic axonal injury (TAI) is still a matter of debate. The present study was conducted to evaluate the effects of moderate, early posttraumatic hypothermia on calpain-mediated spectrin proteolysis (CMSP), implicated in the pathogenesis of TAI. Using moderate (32 degrees C) hypothermia of 90 min duration without rewarming, the density of CMSP immunoreactive/damaged axons was quantified via LM analysis in vulnerable brain stem fiber tracts of hypothermic and normothermic rats subjected to impact acceleration TBI (90 min postinjury survival). To assess the influence of posthypothermic rewarming, a second group of animals was subjected to 90 min of hypothermia followed by 90 min of rewarming to normothermic levels when CMSP was analyzed to detect if any purported CMSP prevention persisted (180 min postinjury survival). Additionally, to determine if this protection translated into comparable cytoskeletal protection in the same foci showing decreased CMSP, antibodies targeting altered/compacted NF subunits were also employed. Moderate hypothermia applied in the acute postinjury period drastically reduced the number of damaged axons displaying CMSP at both time points and significantly reduced NFC immunoreactivity at 180 min postinjury. These results suggest that the neuroprotective effects of hypothermia in TBI are associated with the inhibition of axonal/cytoskeletal damage.


Assuntos
Axônios/enzimologia , Lesões Encefálicas/metabolismo , Calpaína/metabolismo , Citoesqueleto/metabolismo , Hipotermia Induzida , Animais , Anticorpos Monoclonais , Axônios/química , Lesões Encefálicas/terapia , Cálcio/análise , Cálcio/imunologia , Cálcio/metabolismo , Proteínas de Neurofilamentos/análise , Proteínas de Neurofilamentos/imunologia , Proteínas de Neurofilamentos/metabolismo , Tratos Piramidais/química , Tratos Piramidais/citologia , Tratos Piramidais/metabolismo , Ratos , Ratos Sprague-Dawley , Espectrina/análise , Espectrina/imunologia , Espectrina/metabolismo
17.
Acta Neurochir Suppl ; 73: 15-20, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10494336

RESUMO

To gain better insight into the initiating factors involved in traumatically induced axonal injury cats and rats were subjected to various forms of traumatic brain injury. Following injury at intervals ranging from 10 min. to 3 hours, the animals were sacrificed and prepared in accordance with multiple immunocytochemical strategies capable of detecting focal changes in the axolemma, the subaxolemmal spectrin network, the underlying cytoskeleton as well as any related abnormalities in axoplasmic transport. Through these approaches it was recognized that the most severe forms of injury resulted in focal abnormalities of axonal permeability which were observed together with calpain-mediated spectrin proteolysis in the subaxolemmal network. These events were associated with compaction of the underlying neurofilaments and some microtubular loss which occurred without any direct evidence of overt axoplasmic proteolysis with the exception of the most severely injured fibers. In addition to these severely injured axonal profiles, other injured axons did not manifest overt changes in axolemmal permeability or early calpain-mediated spectrin proteolysis but demonstrated dramatic neurofilament and microtubular misalignment and impaired axoplasmic transport. Lastly, other small caliber axons showed another form of intraaxonal change manifested in the local pooling of organelles in the nodal and paranodal regions, with the suggestion that some of these changes may be reversible. In relation to these axonal responses the efficacy of various therapeutic investigations were assessed. The use of calcium chelators showed a trend for protection in those axons manifesting altered axolemmal permeability. However, the use of early and delayed hypothermia demonstrated dramatic protection resulting in significant reduction in the number of damaged axonal profiles. These studies illustrate the diversity and complexity of those axonal responses evoked by traumatic brain injury, suggesting that multiple forms of therapy may be needed to blunt these multifaceted forms of progression.


Assuntos
Axônios/patologia , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Animais , Lesões Encefálicas/tratamento farmacológico , Cálcio/fisiologia , Calpaína/metabolismo , Gatos , Quelantes/uso terapêutico , Progressão da Doença , Microscopia Eletrônica , Fibras Nervosas/fisiologia , Ratos , Espectrina/metabolismo
18.
J Neurotrauma ; 16(6): 511-21, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10391367

RESUMO

Recent observations concerning presumed calcium-induced mitochondrial damage and focal intraaxonal proteolysis in the pathogenesis of traumatic axonal injury (TAI) have opened new perspectives for therapeutic intervention. Studies from our laboratory demonstrated that cyclosporin A (CsA), a potent inhibitor of Ca2+-induced mitochondrial damage, administered 30 min prior to traumatic brain injury preserved mitochondrial integrity in those axonal foci destined to undergo delayed disconnection. We attributed this neuroprotection to the inhibition by CsA of mitochondrial permeability transition (MPT). Additional experiments proved that CsA pretreatment also significantly reduced calcium-induced, calpain-mediated spectrin proteolysis (CMSP) and neurofilament compaction (NFC), pivotal events in the pathogenesis of axonal failure and disconnection. Given these provocative findings the goal of the current study was to evaluate the potential of CsA to inhibit calcium-induced axonal damage in a more clinically relevant postinjury treatment paradigm. To this end, cyclosporin A was administered intrathecally to Sprague Dawley rats 30 min following impact acceleration traumatic brain injury. The first group of animals were sacrificed 120 min postinjury and the density of CMSP and NFC immunoreactive damaged axonal segments of CsA-treated and vehicle-treated injured animals were quantitatively analyzed. A second group of CsA- versus vehicle-treated rats was sacrificed at 24 h postinjury to compare the density of damaged axons displaying beta amyloid precursor protein (APP) immunoreactivity, a signature protein of axonal perturbation and disconnection. Postinjury CsA administration resulted in a significant decrease (>60%) in CMSP/NFC immunoreactivity in corticospinal tracts and medial longitudinal fasciculi. A similar decrease was detected in the density of APP immunoreactive damaged axons, indicating an attenuation of axonal disconnection at 24 h postinjury in CsA-treated animals. These results once again suggest that the maintenance of the functional integrity of the mitochondria can prevent TAI, presumably via the preservation of the local energy homeostasis of the axon. Moreover and perhaps more importantly, these studies also demonstrate the efficacy of CsA administration when given in the early posttraumatic period. Collectively, our findings suggest that a therapeutic window exists for the use of drugs targeting mitochondria and energy regulation in traumatic brain injury.


Assuntos
Axônios/efeitos dos fármacos , Lesões Encefálicas/tratamento farmacológico , Ciclosporina/farmacologia , Fármacos Neuroprotetores/farmacologia , Precursor de Proteína beta-Amiloide/análise , Animais , Axônios/patologia , Biomarcadores/análise , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/metabolismo , Tronco Encefálico/patologia , Cálcio/fisiologia , Calpaína/efeitos dos fármacos , Calpaína/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Masculino , Vias Neurais/lesões , Vias Neurais/fisiopatologia , Proteínas de Neurofilamentos/efeitos dos fármacos , Proteínas de Neurofilamentos/metabolismo , Ratos , Ratos Sprague-Dawley , Espectrina/efeitos dos fármacos , Espectrina/metabolismo
19.
Neuroreport ; 10(2): 353-8, 1999 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-10203334

RESUMO

In traumatic axonal injury, Ca2+ influx across a focally damaged axolemma precipitates local mitochondrial failure, degradation of the subaxolemmal spectrin network and compaction of neurofilaments, which collectively contribute to axonal failure. In previous studies, cyclosporin A pretreatment preserved mitochondrial integrity and attenuated axonal failure following trauma. Here we investigate whether this CsA-linked protection was related to the concomitant blunting of intra-axonal, Ca2+-induced cytoskeletal changes in traumatic axonal injury, assessed with antibodies targeting spectrin proteolysis and neurofilament compaction. CsA pretreatment dramatically reduced Ca2+-induced cytoskeletal damage following injury; CsA-treated rats, compared with vehicle-treated rats, displayed a 70% decrease in immunoreactive/damaged profiles. We suggest that CsA-mediated preservation of mitochondrial integrity enables the restoration of ionic and metabolic homeostasis thereby short-circuiting Ca2+-induced proteolysis in injured axons.


Assuntos
Axônios/efeitos dos fármacos , Lesões Encefálicas/patologia , Cálcio/fisiologia , Ciclosporina/farmacologia , Animais , Axônios/patologia , Biomarcadores , Encéfalo/patologia , Calpaína/fisiologia , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Masculino , Peptídeo Hidrolases/metabolismo , Tratos Piramidais/metabolismo , Ratos , Ratos Sprague-Dawley , Espectrina/metabolismo
20.
J Neuropathol Exp Neurol ; 58(4): 365-75, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10218632

RESUMO

In animals and man, traumatic brain injury (TBI) results in axonal injury (AI) that contributes to morbidity and mortality. Such injured axons show progressive change leading to axonal disconnection. Although several theories implicate calcium in the pathogenesis of AI, experimental studies have failed to confirm its pivotal role. To explore the contribution of Ca2+-induced proteolysis to axonal injury, this study was undertaken in an animal model of TBI employing antibodies targeting both calpain-mediated spectrin proteolysis (CMSP) and focal neurofilament compaction (NFC), a marker of intra-axonal cytoskeletal perturbation, at 15-120 minutes (min) postinjury. Light microscopy (LM) revealed that TBI consistently evoked focal, intra-axonal CMSP that was spatially and temporally correlated with NFC. These changes were seen at 15 min postinjury with significantly increasing number of axons demonstrating CMSP immunoreactivity over time postinjury. Electron microscopy (EM) demonstrated that at 15 min postinjury CMSP was confined primarily to the subaxolemmal network. With increasing survival (30-120 min) CMSP filled the axoplasm proper. These findings provide the first direct evidence for focal CMSP in the pathogenesis of generalized/diffuse AI. Importantly, they also reveal an initial subaxolemmal involvement prior to induction of a more widespread axoplasmic change indicating a spatial-temporal compartmentalization of the calcium-induced proteolytic process that may be amenable to rapid therapeutic intervention.


Assuntos
Axônios/enzimologia , Lesões Encefálicas/metabolismo , Calpaína/metabolismo , Espectrina/metabolismo , Animais , Axônios/química , Axônios/ultraestrutura , Calpaína/análise , Compartimento Celular/fisiologia , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Modelos Animais de Doenças , Microscopia Eletrônica , Proteínas de Neurofilamentos/análise , Proteínas de Neurofilamentos/metabolismo , Ratos , Ratos Sprague-Dawley , Espectrina/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...