Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 9(2): 101, 2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29367701

RESUMO

The complement system is a crucial component of immunity, and its activation has critical roles in neuroinflammatory response and cellular damage following closed head injury (CHI). We previously demonstrated that systemically injected induced neural stem cells (iNSCs) could modulate complement activation to ameliorate neuronal apoptosis in mouse CHI models. However, it remains unknown whether iNSC derivatives can regulate complement activation. In the present study, after CHI mouse serum treatment, we found dramatic decreases in the cellular viabilities of differentiated iNSCs. Interestingly, following CHI mouse serum treatment, the death of astrocytes derived from iNSCs which were pre-treated with CHI mouse serum was significantly decreased. Meanwhile, the deposition of C3 (C3d) and C5b-9 in these astrocytes was substantially reduced. Remarkably, we detected increased expression of complement receptor type 1-related protein y (Crry) in these astrocytes. Moreover, these astrocytes could reduce the numbers of apoptotic neurons via Crry expression post-CHI mouse serum treatment. Additionally, intracerebral-transplanted iNSCs, pre-treated with CHI mouse serum, significantly increased the levels of Crry expression in astrocytes to reduce the accumulation of C3d and C9 and the death of neurons in the brains of CHI mice. In summary, iNSCs receiving CHI mouse serum pre-treatment could enhance the expression of Crry in iNSC-derived astrocytes to modulate complement activation and mediate neuroprotection following CHI.


Assuntos
Astrócitos/citologia , Ativação do Complemento , Traumatismos Cranianos Fechados/imunologia , Traumatismos Cranianos Fechados/prevenção & controle , Células-Tronco Neurais/citologia , Neuroproteção , Animais , Apoptose , Contagem de Células , Diferenciação Celular , Sobrevivência Celular , Proteínas do Sistema Complemento/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/transplante , Receptores de Complemento/metabolismo , Receptores de Complemento 3b , Soro/metabolismo
2.
Stem Cell Res ; 23: 132-142, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28743043

RESUMO

Closed head injury (CHI) usually results in severe and permanent neurological impairments, which are caused by several intertwined phenomena, such as cerebral edema, blood-brain barrier (BBB) disruption, neuronal loss, astroglial scarring and inflammation. We previously reported that induced neural stem cells (iNSCs), similar to neural stem cells (NSCs), can accelerate neurological recovery in vivo and produce neurotrophic factors in vitro. However, the effects of iNSC neurotrophy following CHI were not determined. Moreover, whether iNSCs have immunomodulatory properties is unknown. Mouse models of CHI were established using a standardized weight-drop device and assessed by neurological severity score (NSS). Although these models fail to mimic the complete spectrum of human CHI, they reproduce impairment in neurological function observed in clinical patients. Syngeneic iNSCs or NSCs were separately transplanted into the brains of CHI mice at 12h after CHI. Neurological impairment post-CHI was evaluated by several tests. Animals were sacrificed for morphological and molecular biological analyses. We discovered that iNSC administration promoted neurological functional recovery in CHI mice and reduced cerebral edema, BBB disruption, cell death and astroglial scarring following trauma. Implanted iNSCs could up-regulate brain-derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF) levels to support the survival of existing neurons after CHI. In addition, engrafted iNSCs decreased immune cell recruitment and pro-inflammatory cytokine expression in the brain post-injury. Moreover, we found significant nuclear factor-kappaB (NF-κB) inhibition in the presence of iNSC grafts. In short, iNSCs exert neurotrophic and immunomodulatory effects that mitigate CHI-induced neurological impairment.


Assuntos
Traumatismos Cranianos Fechados/imunologia , Traumatismos Cranianos Fechados/terapia , Imunomodulação , Células-Tronco Neurais/transplante , Transplante de Células-Tronco , Animais , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Edema Encefálico/patologia , Edema Encefálico/fisiopatologia , Sobrevivência Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Gliose/patologia , Gliose/fisiopatologia , Traumatismos Cranianos Fechados/patologia , Traumatismos Cranianos Fechados/fisiopatologia , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Neurônios/metabolismo , Neutrófilos/metabolismo , Fosforilação , Recuperação de Função Fisiológica , Linfócitos T/metabolismo
3.
Georgian Med News ; (263): 111-118, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28452737

RESUMO

This article describes the immunological events and investigates important mediators of the inflammatory immune response after experimental cranial trauma combined with type I diabetes. It was shown that the rats with induced diabetes and traumatic brain injury suffered more profound damage to the immune system that was not restored back to the basal level within 14 days. Proinflammatory cytokine concentrations increased, while anti-inflammatory cytokine concentrations decreased by day 14, increasing risk for systemic inflammatory response syndrome and multi-organ failure. Factors responsible for humoral immunity and cell-mediated immunity were consistently lower by day 14, which might make diabetic rats more susceptible to infections compared to rats with brain injury alone.


Assuntos
Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Tipo 1/imunologia , Traumatismos Cranianos Fechados/imunologia , Animais , Citocinas/sangue , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Tipo 1/complicações , Traumatismos Cranianos Fechados/complicações , Imunidade Celular , Imunidade Humoral , Masculino , Insuficiência de Múltiplos Órgãos/imunologia , Ratos Wistar , Risco , Síndrome de Resposta Inflamatória Sistêmica/imunologia
4.
Sci Rep ; 7: 45989, 2017 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-28383046

RESUMO

Complement activation plays important roles in the pathogenesis of central nervous system (CNS) diseases. Patients face neurological disorders due to the development of complement activation, which contributes to cell apoptosis, brain edema, blood-brain barrier dysfunction and inflammatory infiltration. We previously reported that induced neural stem cells (iNSCs) can promote neurological functional recovery in closed head injury (CHI) animals. Remarkably, we discovered that local iNSC grafts have the potential to modulate CNS inflammation post-CHI. In this study, we aimed to explore the role of systemically delivered iNSCs in complement activation following CNS injury. Our data showed that iNSC grafts decreased the levels of sera C3a and C5a and down-regulated the expression of C3d, C9, active Caspase-3 and Bax in the brain, kidney and lung tissues of CHI mice. Furthermore, iNSC grafts decreased the levels of C3d+/NeuN+, C5b-9+/NeuN+, C3d+/Map2+ and C5b-9+/Map2+ neurons in the injured cortices of CHI mice. Subsequently, we explored the mechanisms underlying these effects. With flow cytometry analysis, we observed a dramatic increase in complement receptor type 1-related protein y (Crry) expression in iNSCs after CHI mouse serum treatment. Moreover, both in vitro and in vivo loss-of-function studies revealed that iNSCs could modulate complement activation via Crry expression.


Assuntos
Ativação do Complemento/imunologia , Traumatismos Cranianos Fechados/imunologia , Traumatismos Cranianos Fechados/terapia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/transplante , Transplante de Células-Tronco , Animais , Barreira Hematoencefálica/patologia , Caspase 3/metabolismo , Proteínas do Sistema Complemento/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Edema/patologia , Traumatismos Cranianos Fechados/sangue , Traumatismos Cranianos Fechados/patologia , Inflamação/patologia , Camundongos , Neurônios/metabolismo , Infiltração de Neutrófilos , Especificidade de Órgãos , Permeabilidade , Receptores de Complemento/metabolismo , Receptores de Complemento 3b , Proteína X Associada a bcl-2/metabolismo
5.
Toxicol Mech Methods ; 26(1): 1-10, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26275125

RESUMO

CONTEXT: Traumatic brain injury in the pediatric population can have a great economic and emotional impact on both the child's family and society. OBJECTIVE: The present study aimed to compare the effects of carnosine (CAR) and/or cyclosporine A (CyA) on oxidative brain damage after closed head injury (CHI) in immature rats. MATERIALS AND METHODS: Thirty-day-old rat pups were divided into five groups: non-traumatic control group, trauma group underwent CHI, trauma group injected with CAR (200 mg/kg, i.p.) following CHI for 7 d, trauma group injected with CyA (20 mg/kg, i.p.) given 15 min and 24 h after CHI, and trauma group treated with CAR and CyA. At the end of the treatment, rats were sacrificed; blood and brains were collected for assessing different biochemical parameters. RESULTS: Trauma significantly increased brain level of malondialdehyde, nitric oxide, glucose, calcium, inflammatory mediators. Brain DNA damage was confirmed by comet assay and the significant increase in brain caspase-3 activity. Moreover, the serum level of Fas ligand in traumatized animals was significantly elevated. Concomitant decrease in brain-reduced glutathione (GSH) and calcium-adenosine triphosphatase activity was observed in the traumatized-untreated group. Treatment of traumatized animals with CAR and/or CyA ameliorated all the biochemical changes induced by CHI with marked protective effect in the combination group. DISCUSSION AND CONCLUSION: CAR and CyA exerted a synergistic neuroprotective effect against CHI through blocking the induction of lipid peroxidation, reducing inflammatory, and oxidative stress biomarkers, preserving brain GSH content, and reducing the alterations in brain apoptotic biomarkers in traumatized animals.


Assuntos
Apoptose/efeitos dos fármacos , Carnosina/uso terapêutico , Ciclosporina/uso terapêutico , Traumatismos Cranianos Fechados/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Animais , Apoptose/imunologia , Biomarcadores/análise , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Encéfalo/imunologia , Encéfalo/metabolismo , Carnosina/administração & dosagem , Ciclosporina/administração & dosagem , Citocinas/imunologia , Sinergismo Farmacológico , Quimioterapia Combinada , Traumatismos Cranianos Fechados/imunologia , Traumatismos Cranianos Fechados/metabolismo , Traumatismos Cranianos Fechados/patologia , Masculino , Fármacos Neuroprotetores/administração & dosagem , Estresse Oxidativo/imunologia , Ratos Wistar
6.
Crit Care Med ; 42(9): 2092-100, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25014065

RESUMO

OBJECTIVES: Traumatic brain injury results in significant morbidity and mortality and is associated with infectious complications, particularly pneumonia. However, whether traumatic brain injury directly impacts the host response to pneumonia is unknown. The objective of this study was to determine the nature of the relationship between traumatic brain injury and the prevalence of pneumonia in trauma patients and investigate the mechanism of this relationship using a murine model of traumatic brain injury with pneumonia. DESIGN: Data from the National Trauma Data Bank and a murine model of traumatic brain injury with postinjury pneumonia. SETTING: Academic medical centers in Cincinnati, OH, and Boston, MA. PATIENTS/SUBJECTS: Trauma patients in the National Trauma Data Bank with a hospital length of stay greater than 2 days, age of at least 18 years at admission, and a blunt mechanism of injury. Subjects were female ICR mice 8-10 weeks old. INTERVENTIONS: Administration of a substance P receptor antagonist in mice. MEASUREMENTS AND MAIN RESULTS: Pneumonia rates were measured in trauma patients before and after risk adjustment using propensity scoring. In addition, survival and pulmonary inflammation were measured in mice undergoing traumatic brain injury with or without pneumonia. After risk adjustment, we found that traumatic brain injury patients had significantly lower rates of pneumonia compared to blunt trauma patients without traumatic brain injury. A murine model of traumatic brain injury reproduced these clinical findings with mice subjected to traumatic brain injury demonstrating increased bacterial clearance and survival after induction of pneumonia. To determine the mechanisms responsible for this improvement, the substance P receptor was blocked in mice after traumatic brain injury. This treatment abrogated the traumatic brain injury-associated increases in bacterial clearance and survival. CONCLUSIONS: The data demonstrate that patients with traumatic brain injury have lower rates of pneumonia compared to non-head-injured trauma patients and suggest that the mechanism of this effect occurs through traumatic brain injury-induced release of substance P, which improves innate immunity to decrease pneumonia.


Assuntos
Lesões Encefálicas/imunologia , Pneumonia/imunologia , Substância P/fisiologia , Ferimentos não Penetrantes/imunologia , Adulto , Fatores Etários , Idoso , Animais , Boston , Lesões Encefálicas/complicações , Feminino , Traumatismos Cranianos Fechados/complicações , Traumatismos Cranianos Fechados/imunologia , Humanos , Escala de Gravidade do Ferimento , Masculino , Camundongos , Camundongos Endogâmicos ICR , Pessoa de Meia-Idade , Antagonistas dos Receptores de Neurocinina-1/farmacologia , Pneumonia/etiologia , Respiração Artificial , Fatores Sexuais , Centros de Traumatologia , Ferimentos não Penetrantes/complicações
7.
J Cereb Blood Flow Metab ; 34(8): 1391-401, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24849666

RESUMO

A mild insult to the brain can sometimes trigger secondary brain injury, causing severe postconcussion syndrome, but the underlying mechanism is ill understood. We show here that secondary brain injury occurs consistently in mice lacking immediate early responsive gene X-1 (IEX-1), after a gentle impact to the head, which closely simulates mild traumatic brain injury in humans. The pathologic lesion was characterized by extensive cell death, widespread leukocyte infiltrates, and severe tissue loss. On the contrary, a similar insult did not induce any secondary injury in wild-type mice. Strikingly, noninvasive exposure of the injured head to a low-level laser at 4 hours after injury almost completely prevented the secondary brain injury in IEX-1 knockout mice. The low-level laser therapy (LLLT) suppressed proinflammatory cytokine expression like interleukin (IL)-1ß and IL-6 but upregulated TNF-α. Moreover, although lack of IEX-1 compromised ATP synthesis, LLLT elevated its production in injured brain. The protective effect of LLLT may be ascribed to enhanced ATP production and selective modulation of proinflammatory mediators. This new closed head injury model provides an excellent tool to investigate the pathogenesis of secondary brain injury as well as the mechanism underlying the beneficial effect of LLLT.


Assuntos
Lesões Encefálicas/prevenção & controle , Genes Precoces , Traumatismos Cranianos Fechados/radioterapia , Proteínas Imediatamente Precoces/deficiência , Terapia com Luz de Baixa Intensidade , Animais , Lesões Encefálicas/genética , Lesões Encefálicas/imunologia , Lesões Encefálicas/patologia , Citocinas/imunologia , Traumatismos Cranianos Fechados/genética , Traumatismos Cranianos Fechados/imunologia , Traumatismos Cranianos Fechados/patologia , Proteínas Imediatamente Precoces/genética , Escala de Gravidade do Ferimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Exame Neurológico , Prevenção Secundária
8.
Neurocrit Care ; 19(1): 125-34, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23269559

RESUMO

BACKGROUND: Use of antiepileptic drugs (AED's) is common in the neurocritical care setting. However, there remains a great deal of controversy regarding the optimal agent. Studies associating the prophylactic use of AED's with poor outcomes are heavily biased by the prevalent use of phenytoin, an agent highly associated with deleterious effects. In the current study, we evaluate lacosamide for neuroprotective properties in a murine model of closed head injury. METHODS: Mice were subjected to moderate closed head injury using a pneumatic impactor, and then treated with either low-dose (6 mg/kg) or high-dose (30 mg/kg) lacosamide or vehicle at 30 min post-injury, and twice daily for 3 days after injury. Motor and cognitive functional assessments were performed following injury using rotarod and Morris Water Maze, respectively. Neuronal injury and microglial activation were measured by flourojade-B, NeuN, and F4/80 staining at 1 and 7 days post-injury. Timm's staining was also performed to assess lacosamide effects on mossy fiber axonal sprouting. To evaluate possible mechanisms of lacosamide effects on the inflammatory response to injury, an RNA expression array was used to evaluate for alterations in differential gene expression patterns in injured mice following lacosamide or vehicle treatments. RESULTS: High-dose lacosamide was associated with improved functional outcome on both the rotarod and Morris Water Maze. High-dose lacosamide was also associated with a reduction of neuronal injury at 24 h post-injury. However, the reduction in neuronal loss observed early did not result in greater neuronal density at 31 days post-injury based on unbiased stereology of NeuN staining. High-dose lacosamide was also associated with a significant reduction in microglial activation at 7 days post-injury. The therapeutic effects of lacosamide are associated with a delay in injury-related changes in RNA expression of a subset of inflammatory mediator genes typically seen at 24 h post-injury. CONCLUSIONS: Administration of lacosamide improves functional performance, and reduces histological evidence of acute neuronal injury and neuroinflammation in a murine model of closed head injury. Lacosamide effects appear to be mediated via a reduction or delay in the acute inflammatory response to injury. Prior clinical and animal studies have found antiepileptic treatment following injury to be detrimental, though these studies are biased by the common use of older medications such as phenytoin. Our current results as well as prior work on levetiracetam suggest the newer AED's may be beneficial in the setting of acute brain injury.


Assuntos
Acetamidas/farmacologia , Anticonvulsivantes/farmacologia , Lesões Encefálicas/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Animais , Lesões Encefálicas/imunologia , Lesões Encefálicas/patologia , Modelos Animais de Doenças , Encefalite/tratamento farmacológico , Encefalite/imunologia , Encefalite/patologia , Traumatismos Cranianos Fechados/tratamento farmacológico , Traumatismos Cranianos Fechados/imunologia , Traumatismos Cranianos Fechados/patologia , Lacosamida , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/patologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Transcriptoma/imunologia , Resultado do Tratamento
9.
J Neurotrauma ; 29(6): 1233-42, 2012 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-22335783

RESUMO

The role of adaptive immunity in contributing to post-traumatic neuroinflammation and neuropathology after head injury remains largely unexplored. The present study was designed to investigate the pathophysiological sequelae of closed head injury in Rag1(-/-) mice devoid of mature B and T lymphocytes. C57BL/6 wild-type and Rag1(-/-) mice were subjected to experimental closed head injury, using a standardized weight-drop device. Outcome parameters consisted of neurological scoring, quantification of blood-brain barrier (BBB) function, measurement of inflammatory markers and mediators of apoptosis in serum and brain tissue, and assessment of neuronal cell death, astrogliosis, and tissue destruction. There was no difference between wild-type and Rag1(-/-) mice with regard to injury severity and neurological impairment for up to 7 days after head injury. The extent of BBB dysfunction was in a similar range for both groups. Quantification of complement activation fragments in serum revealed significantly attenuated C3a levels in Rag1(-/-) mice compared to wild-type animals. In contrast, the levels of pro- and anti-inflammatory cytokines and pro-apoptotic and anti-apoptotic mediators remained in a similar range for both groups, and the histological analysis of brain sections did not reveal a difference in reactive astrogliosis, tissue destruction, and neuronal cell death in Rag1(-/-) compared to wild-type mice. These findings suggest that adaptive immunity is not of crucial importance for initiating and sustaining the inflammatory neuropathology after closed head injury. The attenuated extent of post-traumatic complement activation seen in Rag1(-/-) mice implies a cross-talk between innate and adaptive immune responses, which requires further investigation in future studies.


Assuntos
Imunidade Adaptativa , Genes RAG-1 , Traumatismos Cranianos Fechados/imunologia , Traumatismos Cranianos Fechados/patologia , Animais , Linfócitos B/imunologia , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Western Blotting , Ativação do Complemento/imunologia , Traumatismos Cranianos Fechados/fisiopatologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T/imunologia
10.
Neurobiol Dis ; 40(2): 394-403, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20621186

RESUMO

The contribution of infiltrated neutrophils to secondary damage following traumatic brain injury remains controversial. Chemokines that regulate neutrophil migration by signaling through the CXCR2 receptor are markedly elevated by brain injury and are associated with the propagation of secondary damage. This study thus investigated the function of CXCR2 in posttraumatic inflammation and secondary degeneration by examining Cxcr2-deficient (Cxcr2(-/-)) mice over 14 days following closed head injury (CHI). We demonstrate a significant attenuation of neutrophil infiltration in Cxcr2(-/-) mice at 12 hours and 7 days after CHI, despite increased levels of CXC neutrophil-attracting chemokines in the lesioned cortex. This coincides with reduced tissue damage, neuronal loss, and cell death in Cxcr2(-/-) mice compared to wild-type controls, with heterozygotes showing intermediate responses. In contrast, blood-brain barrier permeability and functional recovery did not appear to be affected by Cxcr2 deletion. This study highlights the deleterious contribution of neutrophils to posttraumatic neurodegeneration and demonstrates the importance of CXC chemokine signaling in this process. Therefore, CXCR2 antagonistic therapeutics currently in development for other inflammatory conditions may also be of benefit in posttraumatic neuroinflammation.


Assuntos
Córtex Cerebral/imunologia , Traumatismos Cranianos Fechados/imunologia , Infiltração de Neutrófilos/imunologia , Receptores de Interleucina-8B/deficiência , Fatores Etários , Animais , Barreira Hematoencefálica/patologia , Morte Celular , Córtex Cerebral/patologia , Quimiocinas/imunologia , Citocinas/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Traumatismos Cranianos Fechados/patologia , Heterozigoto , Homozigoto , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores de Interleucina-8B/genética , Recuperação de Função Fisiológica
11.
Acta Neurochir (Wien) ; 149(3): 281-9, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17288002

RESUMO

BACKGROUND: The contribution of brain edema to brain swelling in cases of traumatic brain injury (TBI) remains a critical problem. We believe that inflammatory reactions may play a fundamental role in brain swelling following a head injury. Although possible roles of microglia activation and the release of mediators have been suggested, direct evidence of cellular immune reactivity in diffuse brain injury following closed head trauma is lacking. Accordingly, the objective of this study was to assess the temporal pattern of microglia activation and lymphocyte migration in an experimental model of TBI. METHOD: An impact acceleration TBI model was utilized to induce diffuse brain damage in adult Wistar rats. The animals were separated into three groups: unoperated controls, sham-operated controls and trauma group. At various times after TBI induction (5 min-24 h), rats were perfused transcardially. Sagittal brain sections were analyzed with immunohistochemical markers of CD3 to reveal the presence of T-lymphocytes, and by immunochemistry for the detection of CD11b to reveal microglia activation within the brain parenchyma. FINDINGS: In the control groups, scattered T-cells were found in the brain parenchyma. In the trauma group, TBI induced microglia activation and a transient biphasic T-cell infiltration of the brain parenchyma in all regions was found, beginning as early as 30 min post injury and reaching its maximum values at 45 min and 3 h after trauma induction. CONCLUSION: These results lead us to suggest that the acute response to severe head trauma with early edema formation is likely to be associated with inflammatory events which might be triggered by activated microglia and infiltrating lymphocytes. It is difficult to overestimate the clinical significance of these observations, as the early and targeted treatment of patients with severe head injuries with immunosuppressive medication may result in a far more favorable outcome.


Assuntos
Lesões Encefálicas/imunologia , Traumatismos Cranianos Fechados/imunologia , Imunidade Celular/imunologia , Aceleração , Animais , Encéfalo/imunologia , Encéfalo/patologia , Edema Encefálico/imunologia , Edema Encefálico/patologia , Lesões Encefálicas/patologia , Antígeno CD11b/análise , Complexo CD3/imunologia , Modelos Animais de Doenças , Traumatismos Cranianos Fechados/patologia , Pressão Intracraniana/fisiologia , Linfocitose/imunologia , Linfocitose/patologia , Masculino , Microglia/imunologia , Microglia/patologia , Ratos , Linfócitos T/imunologia , Linfócitos T/patologia
12.
Brain Res Brain Res Rev ; 48(2): 388-99, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15850678

RESUMO

Closed head injury (CHI) remains the leading cause of death and persisting neurological impairment in young individuals in industrialized nations. Research efforts in the past years have brought evidence that the intracranial inflammatory response in the injured brain contributes to the neuropathological sequelae which are, in large part, responsible for the adverse outcome after head injury. The presence of hypoxia and hypotension in the early resuscitative period of brain-injured patients further aggravates the inflammatory response in the brain due to ischemia/reperfusion-mediated injuries. The profound endogenous neuroinflammatory response after CHI, which is phylogenetically aimed at defending the intrathecal compartment from invading pathogens and repairing lesioned brain tissue, contributes to the development of cerebral edema, breakdown of the blood-brain barrier, and ultimately to delayed neuronal cell death. However, aside from these deleterious effects, neuroinflammation has been recently shown to mediate neuroreparative mechanisms after brain injury as well. This "dual effect" of neuroinflammation was the focus of extensive experimental and clinical research in the past years and has lead to an expanded basic knowledge on the cellular and molecular mechanisms which regulate the intracranial inflammatory response after CHI. Thus, head injury has recently evolved as an inflammatory and immunological disease much more than a pure traumatological, neurological, or neurosurgical entity. The present review will summarize the so far known mechanisms of posttraumatic neuroinflammation after CHI, based on data from clinical and experimental studies, with a special focus on the role of pro-inflammatory cytokines, chemokines, and the complement system.


Assuntos
Traumatismos Cranianos Fechados/complicações , Traumatismos Cranianos Fechados/imunologia , Inflamação/complicações , Inflamação/imunologia , Animais , Quimiocinas/metabolismo , Citocinas/metabolismo , Humanos
13.
J Cereb Blood Flow Metab ; 24(10): 1110-8, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15529011

RESUMO

The potential role of the chemokine Fractalkine (CX3CL1) in the pathophysiology of traumatic brain injury (TBI) was investigated in patients with head trauma and in mice after experimental cortical contusion. In control individuals, soluble (s)Fractalkine was present at low concentrations in cerebrospinal fluid (CSF) (12.6 to 57.3 pg/mL) but at much higher levels in serum (21,288 to 74,548 pg/mL). Elevation of sFractalkine in CSF of TBI patients was observed during the whole study period (means: 29.92 to 535.33 pg/mL), whereas serum levels remained within normal ranges (means: 3,100 to 59,159 pg/mL). Based on these differences, a possible passage of sFractalkine from blood to CSF was supported by the strong correlation between blood-brain barrier dysfunction (according to the CSF-/serum-albumin quotient) and sFractalkine concentrations in CSF (R = 0.706; P < 0.01). In the brain of mice subjected to closed head injury, neither Fractalkine protein nor mRNA were found to be augmented; however, Fractalkine receptor (CX3CR1) mRNA steadily increased peaking at 1 week postinjury (P < 0.05, one-way analysis of variance). This possibly implies the receptor to be the key factor determining the action of constitutively expressed Fractalkine. Altogether, these data suggest that the Fractalkine-CX3CR1 protein system may be involved in the inflammatory response to TBI, particularly for the accumulation of leukocytes in the injured parenchyma.


Assuntos
Lesões Encefálicas/metabolismo , Quimiocinas CX3C/líquido cefalorraquidiano , Traumatismos Cranianos Fechados/metabolismo , Proteínas de Membrana/líquido cefalorraquidiano , Adolescente , Adulto , Animais , Barreira Hematoencefálica , Lesões Encefálicas/imunologia , Receptor 1 de Quimiocina CX3C , Quimiocina CX3CL1 , Quimiocinas CX3C/sangue , Quimiocinas CX3C/genética , Modelos Animais de Doenças , Feminino , Traumatismos Cranianos Fechados/imunologia , Humanos , Leucócitos/imunologia , Masculino , Proteínas de Membrana/sangue , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Receptores de Citocinas/genética , Receptores de HIV/genética , Solubilidade
14.
J Cereb Blood Flow Metab ; 23(9): 1070-4, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12973023

RESUMO

The role of intracerebral complement activation after traumatic brain injury remains unclear. In this study, the authors demonstrate that transgenic mice with astrocyte-targeted expression of the soluble complement inhibitor sCrry have a significantly reduced neurologic impairment and improved blood-brain barrier function after closed head injury compared with wild-type C57BL/6 littermates. This work further implicates the complement system as a participant in secondary progression of brain damage after head trauma and provides a strong rationale for future studies of posttraumatic pharmacologic complement inhibition.


Assuntos
Sistema Nervoso Central/imunologia , Ativação do Complemento , Proteínas do Sistema Complemento/imunologia , Traumatismos Cranianos Fechados/fisiopatologia , Fármacos Neuroprotetores/metabolismo , Receptores de Complemento/metabolismo , Animais , Comportamento Animal/fisiologia , Barreira Hematoencefálica/fisiologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Traumatismos Cranianos Fechados/imunologia , Traumatismos Cranianos Fechados/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Complemento/genética , Receptores de Complemento 3b
15.
J Neurotrauma ; 20(6): 559-69, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12906740

RESUMO

Closed head injury often has a devastating outcome, partly because the insult, like other injuries to the central nervous system (CNS), triggers self-destructive processes. During studies of the response to other CNS insults, it was unexpectedly discovered that the immune system, if well controlled, provides protection against self-destructive activities. Here we show that in mice with closed head injury, the immune system plays a key role in the spontaneous recovery. Strain-related differences were observed in the ability to harness a T cell-dependent protective mechanism against the effects of the injury. We further show that the trauma-induced deficit could be reduced, both functionally and anatomically, by post-traumatic vaccination with Cop-1, a synthetic copolymer used to treat patients with multiple sclerosis and found (using a different treatment protocol) to effectively counteract the loss of neurons caused by axonal injury or glutamate-induced toxicity. We suggest that a compound such as Cop-1 can be safely developed as a therapeutic vaccine to boost the body's immune repair mechanisms, thereby providing multifactorial protection against the consequences of brain trauma.


Assuntos
Complexo I de Proteína do Envoltório/uso terapêutico , Traumatismos Cranianos Fechados/tratamento farmacológico , Traumatismos Cranianos Fechados/imunologia , Vacinação/métodos , Animais , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Especificidade da Espécie
16.
J Neuroimmunol ; 109(2): 164-72, 2000 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-10996218

RESUMO

The anaphylatoxin C5a is a potent mediator of inflammation in the CNS. We analyzed the intracerebral expression of the C5a receptor (C5aR) in a model of closed head injury (CHI) in mice. Up-regulation of C5aR mRNA and protein expression was observed mainly on neurons in sham-operated and head-injured wild-type mice at 24 h. In contrast, in TNF/lymphotoxin-alpha knockout mice, the intracerebral C5aR expression remained at low constitutive levels after sham operation, whereas it strongly increased in response to trauma between 24 and 72 h. Interestingly, by 7 days after CHI, the intrathecal C5aR expression was clearly attenuated in the knockout animals. These data show that the posttraumatic neuronal expression of the C5aR is, at least in part, regulated by TNF and lymphotoxin-alpha at 7 days after trauma.


Assuntos
Antígenos CD/genética , Traumatismos Cranianos Fechados/imunologia , Linfotoxina-alfa/genética , Receptores de Complemento/genética , Fator de Necrose Tumoral alfa/genética , Animais , Antígenos CD/análise , Antígenos CD/imunologia , Química Encefálica/imunologia , Expressão Gênica/imunologia , Traumatismos Cranianos Fechados/fisiopatologia , Hibridização In Situ , Linfotoxina-alfa/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/análise , Receptor da Anafilatoxina C5a , Receptores de Complemento/análise , Receptores de Complemento/imunologia , Fator de Necrose Tumoral alfa/imunologia
17.
J Cereb Blood Flow Metab ; 20(2): 369-80, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10698075

RESUMO

Cytokines are important mediators of intracranial inflammation following traumatic brain injury (TBI). In the present study, the neurological impairment and mortality, blood-brain barrier (BBB) function, intracranial polymorphonuclear leukocyte (PMN) accumulation, and posttraumatic neuronal cell death were monitored in mice lacking the genes for tumor necrosis factor (TNF)/lymphotoxin-alpha (LT-alpha) (TNF/LT-alpha-/-) and interleukin-6 (IL-6) and in wild-type (WT) littermates subjected to experimental closed head injury (total n = 107). The posttraumatic mortality was significantly increased in TNF/LT-alpha-/- mice (40%; P < 0.02) compared with WT animals (10%). The IL-6-/- mice also showed a higher mortality (17%) than their WT littermates (5.6%), but the difference was not statistically significant (P > 0.05). The neurological severity score was similar among all groups from 1 to 72 hours after trauma, whereas at 7 days, the TNF/LT-alpha-/- mice showed a tendency toward better neurological recovery than their WT littermates. Interestingly, neither the degree of BBB dysfunction nor the number of infiltrating PMNs in the injured hemisphere was different between WT and cytokine-deficient mice. Furthermore, the analysis of brain sections by in situ DNA nick end labeling (TUNEL histochemistry) at 24 hours and 7 days after head injury revealed a similar extent of posttraumatic intracranial cell death in all animals. These results show that the pathophysiological sequelae of TBI are not significantly altered in mice lacking the genes for the proinflammatory cytokines TNF, LT-alpha, and IL-6. Nevertheless, the increased posttraumatic mortality in TNF/LT-alpha-deficient mice suggests a protective effect of these cytokines by mechanisms that have not been elucidated yet.


Assuntos
Barreira Hematoencefálica/fisiologia , Citocinas/genética , Traumatismos Cranianos Fechados/imunologia , Traumatismos Cranianos Fechados/fisiopatologia , Neutrófilos/imunologia , Animais , Morte Celular , Modelos Animais de Doenças , Traumatismos Cranianos Fechados/mortalidade , Marcação In Situ das Extremidades Cortadas , Interleucina-6/genética , Linfotoxina-alfa/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Exame Neurológico , Neurônios/citologia , Fator de Necrose Tumoral alfa/genética
18.
Acta Neurochir Suppl ; 71: 212-4, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9779187

RESUMO

UNLABELLED: Previous studies in our laboratory have shown that controlled cortical impact (CCI) produces an acute inflammatory response in rat brain, including neutrophil accumulation and upregulation of cell adhesion molecules. The purpose of this study was to compare the time course of acute inflammation to blood-brain barrier (BBB) breakdown after (CCI) in rats. METHODS: Male Wistar rats (n = 4-7/group) were subjected to CCI (2.5 mm depth, 4 m/s) and injected with Evans-blue dye (2%, 5 ml/kg) at 30 min, 3.5 h, 7.5 h, or 23.5 h after trauma. 30 min after dye injection rats were saline-perfused. BBB permeability was measured by spectrophotometric quantitation of Evans-blue in injured brain. Alternate cryostat sections from the anterior segment of the injured hemisphere were analyzed immunohistochemically for neutrophils (MoAb RP-3 vs rat neutrophils) or E-selectin (MoAb vs E-selectin). Neutrophils and E-selectin-positive blood vessels were quantitated by light microscopy in 100x cortical and hippocampal fields. RESULTS AND CONCLUSIONS: BBB breakdown was maximal early after CCI, whereas maximum E-selectin upregulation (8 h) and neutrophil accumulation (24 h) occurred later. Events other than acute inflammation initiate BBB permeability after CCI. Acute inflammation may contribute to BBB permeability at 4 h to 24 h after CCI.


Assuntos
Barreira Hematoencefálica/imunologia , Córtex Cerebral/lesões , Selectina E/metabolismo , Traumatismos Cranianos Fechados/imunologia , Inflamação Neurogênica/imunologia , Neutrófilos/imunologia , Animais , Permeabilidade da Membrana Celular/imunologia , Córtex Cerebral/imunologia , Masculino , Ratos , Ratos Wistar
19.
Neuroimmunomodulation ; 5(3-4): 143-59, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9730680

RESUMO

Accumulating evidence during the last decade has shown that the CNS can mount a well-defined inflammatory reaction to a variety of insults including trauma, ischemia, transplantation, viral infections as well as neurodegeneration. Many aspects of this centrally derived inflammatory response parallel to some extent the nature of such a reaction in the periphery. Through the recent application of molecular genetic techniques including PCR, utilization of cDNA probes in conjuncture with the availability of highly specific antibodies, new concepts are rapidly emerging as to the molecular mechanisms associated with the development of brain injury. In particular, the importance of cytokines, especially TNFalpha and IL-1beta, is emphasized in the propagation and maintenance of a CNS inflammatory response. This review summarizes evidence in support of a case for ischemia and trauma eliciting an inflammatory condition in the injured brain. The inflammatory condition consists of cells (neutrophils early after the onset of brain injury and subsequently monocyte infiltration) and mediators (cytokines, chemokines and adhesion molecules). It is clear that de novo up-regulation of pro-inflammatory cytokines, chemokines and endothelial-leukocyte adhesion molecules in the brain occurs soon following focal ischemia and trauma and at a time when the tissue injury is evolving. The significance of the inflammatory response and its contribution to brain injury are now becoming better understood. Evidence has emerged in support of the role of cytokines in driving the inflammatory response and that this process is causally related to the degree of brain injury. Evidence reviewed includes: (1) the capacity of specific cytokines to exacerbate brain damage; (2) the capacity of specific cytokine blockade to reduce ischemic brain damage; (3) depletion of circulating neutrophils reduces ischemic brain injury, and (4) antagonists of the endothelial-leukocyte adhesion interactions (e.g. anti-ICAM-1) reduce ischemic brain injury. Targeting the cytokines that drive the brain inflammatory response to injury provides opportunities to intervene with novel therapeutics in stroke and neurotrauma.


Assuntos
Lesões Encefálicas/imunologia , Transtornos Cerebrovasculares/imunologia , Citocinas/imunologia , Traumatismos Cranianos Fechados/imunologia , Lesões Encefálicas/patologia , Transtornos Cerebrovasculares/patologia , Traumatismos Cranianos Fechados/patologia , Humanos
20.
J Head Trauma Rehabil ; 13(1): 11-27, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9565701

RESUMO

The severe hypermetabolism and hypercatabolism seen in patients with severe head injuries results in malnutrition that occurs very rapidly and can cause impaired healing and an increased tendency to infection and multiple organ failure. Thus, early adequate nutritional support plays a role in functional outcome. Total enteral nutrition (TEN) is preferred over total parenteral nutrition (TPN), but TPN should be supplied promptly while increasing TEN to a goal of at least 25 to 35 nonprotein kcal/kg/d and 2.0 to 2.5 g protein/kg/d. Nutritional formulas high in branched chain amino acids, glutamine, arginine, vitamins E and C, and zinc may also have some advantages. Growth hormone may improve anabolism. Hyperglycemia, especially glucose levels exceeding 200 mg/dL, must be prevented and/or treated promptly with insulin or decreased glucose intake. Careful monitoring with indirect calorimetry and nitrogen balance studies should help prevent inadequate protein or excessive carbohydrate intake.


Assuntos
Traumatismos Cranianos Fechados/metabolismo , Traumatismos Cranianos Fechados/terapia , Apoio Nutricional , Reação de Fase Aguda/metabolismo , Nutrição Enteral , Escala de Coma de Glasgow , Traumatismos Cranianos Fechados/imunologia , Humanos , Hiperglicemia/etiologia , Nutrição Parenteral Total
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...