Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Neuropharmacology ; 110(Pt B): 654-659, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25963414

RESUMO

Impact to the head or rapid head acceleration-deceleration can cause traumatic brain injury (TBI) with a characteristic pathology of traumatic axonal injury (TAI) and secondary damage in white matter tracts. Myelin and oligodendrocyte lineage cells have significant roles in the progression of white matter pathology after TBI and in the potential for plasticity and subsequent recovery. The myelination pattern of specific brain regions, such as frontal cortex, may also increase susceptibility to neurodegeneration and psychiatric symptoms after TBI. White matter pathology after TBI depends on the extent and distribution of axon damage, microhemorrhages and/or neuroinflammation. TAI occurs in a pattern of damaged axons dispersed among intact axons in white matter tracts. TAI accompanied by bleeding and/or inflammation produces focal regions of overt tissue destruction, resulting in loss of both axons and myelin. White matter regions with TAI may also exhibit demyelination of intact axons. Demyelinated axons that remain viable have the potential for remyelination and recovery of function. Indeed, animal models of TBI have demonstrated demyelination that is associated with evidence of remyelination, including oligodendrocyte progenitor cell proliferation, generation of new oligodendrocytes, and formation of thinner myelin. Changes in neuronal activity that accompany TBI may also involve myelin remodeling, which modifies conduction efficiency along intact myelinated fibers. Thus, effective remyelination and myelin remodeling may be neurobiological substrates of plasticity in neuronal circuits that require long-distance communication. This perspective integrates findings from multiple contexts to propose a model of myelin and oligodendrocyte lineage cell relevance in white matter injury after TBI. This article is part of the Special Issue entitled 'Oligodendrocytes in Health and Disease'.


Assuntos
Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/fisiopatologia , Plasticidade Celular/fisiologia , Oligodendroglia/patologia , Substância Branca/patologia , Animais , Humanos , Bainha de Mielina/patologia
2.
Exp Neurol ; 275 Pt 3: 328-333, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25697845

RESUMO

Impact-acceleration forces to the head cause traumatic brain injury (TBI) with damage in white matter tracts comprised of long axons traversing the brain. White matter injury after TBI involves both traumatic axonal injury (TAI) and myelin pathology that evolves throughout the post-injury time course. The axon response to initial mechanical forces and secondary insults follows the process of Wallerian degeneration, which initiates as a potentially reversible phase of intra-axonal damage and proceeds to an irreversible phase of axon fragmentation. Distal to sites of axon disconnection, myelin sheaths remain for prolonged periods, which may activate neuroinflammation and inhibit axon regeneration. In addition to TAI, TBI can cause demyelination of intact axons. These evolving features of axon and myelin pathology also represent opportunities for repair. In experimental TBI, demyelinated axons exhibit remyelination, which can serve to both protect axons and facilitate recovery of function. Myelin remodeling may also contribute to neuroplasticity. Efficient clearance of myelin debris is a potential target to attenuate the progression of chronic pathology. During the early phase of Wallerian degeneration, interventions that prevent the transition from reversible damage to axon disconnection warrant the highest priority, based on the poor regenerative capacity of axons in the CNS. Clinical evaluation of TBI will need to address the challenge of accurately detecting the extent and stage of axon damage. Distinguishing the complex white matter changes associated with axons and myelin is necessary for interpreting advanced neuroimaging approaches and for identifying a broader range of therapeutic opportunities to improve outcome after TBI.


Assuntos
Axônios/patologia , Lesões Encefálicas/patologia , Bainha de Mielina/patologia , Substância Branca/patologia , Animais , Axônios/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas/metabolismo , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Humanos , Bainha de Mielina/metabolismo , Substância Branca/metabolismo
3.
J Neuropathol Exp Neurol ; 74(3): 218-32, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25668562

RESUMO

White matter tracts are highly vulnerable to damage from impact-acceleration forces of traumatic brain injury (TBI). Mild TBI is characterized by a low density of traumatic axonal injury, whereas associated myelin pathology is relatively unexplored. We examined the progression of white matter pathology in mice after mild TBI with traumatic axonal injury localized in the corpus callosum. Adult mice received a closed-skull impact and were analyzed from 3 days to 6 weeks post-TBI/sham surgery. At all times post-TBI, electron microscopy revealed degenerating axons distributed among intact fibers in the corpus callosum. Intact axons exhibited significant demyelination at 3 days followed by evidence of remyelination at 1 week. Accordingly, bromodeoxyuridine pulse-chase labeling demonstrated the generation of new oligodendrocytes, identified by myelin proteolipid protein messenger RNA expression, at 3 days post-TBI. Overall oligodendrocyte populations, identified by immunohistochemical staining for CC1 and/or glutathione S-transferase pi, were similar between TBI and sham mice by 2 weeks. Excessively long myelin figures, similar to redundant myelin sheaths, were a significant feature at all post-TBI time points. At 6 weeks post-TBI, microglial activation and astrogliosis were localized to areas of axon and myelin pathology. These studies show that demyelination, remyelination, and excessive myelin are components of white matter degeneration and recovery in mild TBI with traumatic axonal injury.


Assuntos
Lesões Encefálicas/patologia , Progressão da Doença , Bainha de Mielina/patologia , Substância Branca/patologia , Animais , Axônios/patologia , Axônios/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/ultraestrutura , Substância Branca/ultraestrutura
4.
ASN Neuro ; 6(5)2014.
Artigo em Inglês | MEDLINE | ID: mdl-25290062

RESUMO

The regenerative capacity of the central nervous system must be optimized to promote repair following traumatic brain injury (TBI) and may differ with the site and form of damage. Sonic hedgehog (Shh) maintains neural stem cells and promotes oligodendrogenesis. We examined whether Shh signaling contributes to neuroblast (doublecortin) or oligodendrocyte progenitor (neural/glial antigen 2 [NG2]) responses in two distinct TBI models. Shh-responsive cells were heritably labeled in vivo using Gli1-CreER(T2);R26-YFP bitransgenic mice with tamoxifen administration on Days 2 and 3 post-TBI. Injury to the cerebral cortex was produced with mild controlled cortical impact. Yellow fluorescent protein (YFP) cells decreased in cortical lesions. Total YFP cells increased in the subventricular zone (SVZ), indicating Shh pathway activation in SVZ cells, including doublecortin-labeled neuroblasts. The alternate TBI model produced traumatic axonal injury in the corpus callosum. YFP cells decreased within the SVZ and were rarely double labeled as NG2 progenitors. NG2 progenitors increased in the cortex, with a similar pattern in the corpus callosum. To further test the potential of NG2 progenitors to respond through Shh signaling, Smoothened agonist was microinjected into the corpus callosum to activate Shh signaling. YFP cells and NG2 progenitors increased in the SVZ but were not double labeled. This result indicates that either direct Smoothened activation in NG2 progenitors does not signal through Gli1 or that Smoothened agonist acts indirectly to increase NG2 progenitors. Therefore, in all conditions, neuroblasts exhibited differential Shh pathway utilization compared with oligodendrocyte progenitors. Notably, cortical versus white matter damage from TBI produced opposite responses of Shh-activated cells within the SVZ.


Assuntos
Lesões Encefálicas/patologia , Córtex Cerebral/patologia , Ventrículos Cerebrais/patologia , Proteínas Hedgehog/metabolismo , Oligodendroglia/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Substância Branca/patologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Diferenciação Celular , Cicloexilaminas/farmacologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Proteínas Hedgehog/genética , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Microinjeções , Proteínas do Tecido Nervoso/metabolismo , Tiofenos/farmacologia , Transdução Genética , Proteína GLI1 em Dedos de Zinco
5.
Neurosci Lett ; 548: 280-5, 2013 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-23684572

RESUMO

In demyelinating diseases, such as multiple sclerosis, remyelination offers the potential to recover function of viable denuded axons by restoring saltatory conduction and/or protecting from further damage. Mice with genetic reduction of fibroblast growth factor 2 (Fgf2) or Fgf receptor 1 (Fgfr1) exhibit dramatically improved remyelination following experimental demyelination with cuprizone. The current studies are the first to test neurobehavioral outcomes with these gene deletions that improved remyelination. The cuprizone protocols used did not produce overt abnormalities but did reduce bilateral sensorimotor coordination (complex wheel task) and increase sociability (two chamber apparatus with novel mouse). A significant effect of genotype was observed on the complex wheel task but not in the sociability apparatus. Specifically, complex wheel velocities for Fgf2 nulls improved significantly after removal of cuprizone from the diet. This improvement in Fgf2 null mice occurred following either acute (6 weeks) or chronic (12 weeks) demyelination. Plp/CreERT:Fgfr1(fl/fl) mice administered tamoxifen at 10 weeks of cuprizone treatment to induce Fgfr1 knockdown also showed improved recovery of running velocities on the complex wheels. Therefore, constitutive deletion of Fgf2 or Fgfr1 knockdown in oligodendrocyte lineage cells is sufficient to overcome impairment of sensorimotor coordination after cuprizone demyelination.


Assuntos
Comportamento Animal/fisiologia , Doenças Desmielinizantes/fisiopatologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Relações Interpessoais , Fibras Nervosas Mielinizadas/metabolismo , Desempenho Psicomotor/fisiologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Cuprizona , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/patologia , Fator 2 de Crescimento de Fibroblastos/genética , Camundongos , Camundongos Transgênicos , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Fibras Nervosas Mielinizadas/patologia , Desempenho Psicomotor/efeitos dos fármacos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Recuperação de Função Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/fisiologia , Transdução de Sinais/efeitos dos fármacos
6.
J Neuropathol Exp Neurol ; 72(12): 1106-25, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24226267

RESUMO

Traumatic brain injury frequently causes traumatic axonal injury (TAI) in white matter tracts. Experimental TAI in the corpus callosum of adult mice was used to examine the effects on oligodendrocyte lineage cells and myelin in conjunction with neuroimaging. The injury targeted the corpus callosum over the subventricular zone, a source of neural stem/progenitor cells. Traumatic axonal injury was produced in the rostral body of the corpus callosum by impact onto the skull at the bregma. During the first week after injury, magnetic resonance diffusion tensor imaging showed that axial diffusivity decreased in the corpus callosum and that corresponding regions exhibited significant axon damage accompanied by hypertrophic microglia and reactive astrocytes. Oligodendrocyte progenitor proliferation increased in the subventricular zone and corpus callosum. Oligodendrocytes in the corpus callosum shifted toward upregulation of myelin gene transcription. Plp/CreER(T):R26IAP reporter mice showed normal reporter labeling of myelin sheaths 0 to 2 days after injury but labeling was increased between 2 and 7 days after injury. Electron microscopy revealed axon degeneration, demyelination, and redundant myelin figures. These findings expand the cell types and responses to white matter injuries that inform diffusion tensor imaging evaluation and identify pivotal white matter changes after TAI that may affect axon vulnerability vs. recovery after brain injury.


Assuntos
Células-Tronco Adultas/fisiologia , Ventrículos Cerebrais/patologia , Corpo Caloso/patologia , Lesão Axonal Difusa/patologia , Oligodendroglia/fisiologia , Animais , Antígenos/metabolismo , Proliferação de Células , Corpo Caloso/citologia , Corpo Caloso/metabolismo , Estudos Transversais , Lesão Axonal Difusa/fisiopatologia , Imagem de Difusão por Ressonância Magnética , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/metabolismo , Estudos Longitudinais , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Fibras Nervosas Mielinizadas/patologia , Oligodendroglia/ultraestrutura , Proteoglicanas/metabolismo , Receptores de Estrogênio/genética , Fatores de Tempo
7.
J Comp Neurol ; 518(14): 2841-53, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20506478

RESUMO

The consequences of dysmyelination are poorly understood and vary widely in severity. The shaking mouse, a quaking allele, is characterized by severe central nervous system (CNS) dysmyelination and demyelination, a conspicuous action tremor, and seizures in approximately 25% of animals, but with normal muscle strength and a normal lifespan. In this study we compare this mutant with other dysmyelinated mutants including the ceramide sulfotransferase deficient (CST-/-) mouse, which are more severely affected behaviorally, to determine what might underlie the differences between them with respect to behavior and longevity. Examination of the paranodal junctional region of CNS myelinated fibers shows that "transverse bands," a component of the junction, are present in nearly all shaking paranodes but in only a minority of CST-/- paranodes. The number of terminal loops that have transverse bands within a paranode and the number of transverse bands per unit length are only moderately reduced in the shaking mutant, compared with controls, but markedly reduced in CST-/- mice. Immunofluorescence studies also show that although the nodes of the shaking mutant are somewhat longer than normal, Na(+) and K(+) channels remain separated, distinguishing this mutant from CST-/- mice and others that lack transverse bands. We conclude that the essential difference between the shaking mutant and others more severely affected is the presence of transverse bands, which serve to stabilize paranodal structure over time as well as the organization of the axolemmal domains, and that differences in the prevalence of transverse bands underlie the marked differences in progressive neurological impairment and longevity among dysmyelinated mouse mutants.


Assuntos
Envelhecimento/fisiologia , Axônios/fisiologia , Doenças Desmielinizantes/fisiopatologia , Fibras Nervosas Mielinizadas/fisiologia , Nós Neurofibrosos/fisiologia , Envelhecimento/patologia , Animais , Axônios/patologia , Axônios/ultraestrutura , Doenças Desmielinizantes/patologia , Camundongos , Camundongos Knockout , Camundongos Mutantes , Atividade Motora/fisiologia , Bainha de Mielina/patologia , Bainha de Mielina/fisiologia , Bainha de Mielina/ultraestrutura , Fibras Nervosas Mielinizadas/patologia , Nervo Óptico/patologia , Nervo Óptico/fisiopatologia , Nervo Óptico/ultraestrutura , Canais de Potássio/metabolismo , Nós Neurofibrosos/patologia , Nós Neurofibrosos/ultraestrutura , Canais de Sódio/metabolismo , Medula Espinal/patologia , Medula Espinal/fisiopatologia , Medula Espinal/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA