Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Bases de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Glia ; 69(5): 1268-1280, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33417729

RESUMEN

Injury to the developing brain during the perinatal period often causes hypomyelination, leading to clinical deficits for which there is an unmet therapeutic need. Dysregulation of inflammation and microglia have been implicated, yet the molecular mechanisms linking these to hypomyelination are unclear. Using human infant cerebrospinal fluid (CSF) and postmortem tissue, we found that microglial activation of the pro-inflammatory molecular complex the NLRP3 inflammasome is associated with pathology. By developing a novel mouse brain explant model of microglial inflammasome activation, we demonstrate that blocking the inflammasome rescues myelination. In human and mouse, we discovered a link between the inflammasome product IL1ß and increased levels of follistatin, an endogenous inhibitor of activin-A. Follistatin treatment was sufficient to reduce myelination, whereas myelination was rescued in injured explants upon follistatin neutralization or supplementation with exogenous activin-A. Our data reveal that inflammasome activation in microglia drives hypomyelination and identifies novel therapeutic strategies to reinstate myelination following developmental injury.


Asunto(s)
Lesiones Encefálicas , Sustancia Blanca , Activinas , Animales , Modelos Animales de Enfermedad , Folistatina , Inflamasomas/metabolismo , Ratones , Microglía/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Sustancia Blanca/metabolismo
2.
Acta Neuropathol ; 135(6): 887-906, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29397421

RESUMEN

The most prevalent neurological disorders of myelin include perinatal brain injury leading to cerebral palsy in infants and multiple sclerosis in adults. Although these disorders have distinct etiologies, they share a common neuropathological feature of failed progenitor differentiation into myelin-producing oligodendrocytes and lack of myelin, for which there is an unmet clinical need. Here, we reveal that a molecular pathology common to both disorders is dysregulation of activin receptors and that activin receptor signaling is required for the majority of myelin generation in development and following injury. Using a constitutive conditional knockout of all activin receptor signaling in oligodendrocyte lineage cells, we discovered this signaling to be required for myelination via regulation of oligodendrocyte differentiation and myelin compaction. These processes were found to be dependent on the activin receptor subtype Acvr2a, which is expressed during oligodendrocyte differentiation and axonal ensheathment in development and following myelin injury. During efficient myelin regeneration, Acvr2a upregulation was seen to coincide with downregulation of Acvr2b, a receptor subtype with relatively higher ligand affinity; Acvr2b was shown to be dispensable for activin receptor-driven oligodendrocyte differentiation and its overexpression was sufficient to impair the abovementioned ligand-driven responses. In actively myelinating or remyelinating areas of human perinatal brain injury and multiple sclerosis tissue, respectively, oligodendrocyte lineage cells expressing Acvr2a outnumbered those expressing Acvr2b, whereas in non-repairing lesions Acvr2b+ cells were increased. Thus, we propose that following human white matter injury, this increase in Acvr2b expression would sequester ligand and consequently impair Acvr2a-driven oligodendrocyte differentiation and myelin formation. Our results demonstrate dysregulated activin receptor signaling in common myelin disorders and reveal Acvr2a as a novel therapeutic target for myelin generation following injury across the lifespan.


Asunto(s)
Receptores de Activinas/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Oligodendroglía/metabolismo , Receptores de Activinas/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Células Cultivadas , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Oligodendroglía/patología , Ratas Sprague-Dawley , Técnicas de Cultivo de Tejidos , Andamios del Tejido
3.
Nat Neurosci ; 22(7): 1046-1052, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31182869

RESUMEN

Failed regeneration of CNS myelin contributes to clinical decline in neuroinflammatory and neurodegenerative diseases, for which there is an unmet therapeutic need. Here we reveal that efficient remyelination requires death of proinflammatory microglia followed by repopulation to a pro-regenerative state. We propose that impaired microglia death and/or repopulation may underpin dysregulated microglia activation in neurological diseases, and we reveal therapeutic targets to promote white matter regeneration.


Asunto(s)
Enfermedades Desmielinizantes/fisiopatología , Microglía/fisiología , Regeneración Nerviosa/fisiología , Animales , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/patología , Enfermedades Desmielinizantes/inducido químicamente , Femenino , Perfilación de la Expresión Génica , Humanos , Inflamación , Lisofosfatidilcolinas/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/clasificación , Esclerosis Múltiple/patología , Necrosis , Nestina/análisis , Fagocitosis , Ratas , Ratas Sprague-Dawley , Análisis de Secuencia de ARN , Sustancia Blanca/fisiología
4.
Front Physiol ; 9: 1299, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30258368

RESUMEN

Background: Preterm birth is the leading risk factor for perinatal white matter injury, which can lead to motor and neuropsychiatric impairment across the life course. There is an unmet clinical need for therapeutics. White matter injury is associated with an altered inflammatory response in the brain, primarily led by microglia, and subsequent hypomyelination. However, microglia can release both damaging and trophic factors in response to injury, and a comprehensive assessment of these factors in the preterm central nervous system (CNS) has not been carried out. Method: A custom antibody array was used to assess relative levels of 50 inflammation- and myelination-associated proteins in the cerebrospinal fluid (CSF) of preterm infants in comparison to term controls. Results: Fifteen proteins differed between the groups: BDNF, BTC, C5a, FasL, Follistatin, IL-1ß, IL-2, IL-4, IL-9, IL-17A, MIP-1α, MMP8, SPP1, TGFß, and TNFß (p < 0.05). To investigate the temporal regulation of these proteins after injury, we mined a gene expression dataset of microglia isolated from a mouse model of developmental white matter injury. Microglia in the experimental model showed dynamic temporal expression of genes encoding these proteins, with an initial and sustained pro-inflammatory response followed by a delayed anti-inflammatory response, and a continuous expression of genes predicted to inhibit healthy myelination. Conclusion: Preterm CSF shows a distinct neuroinflammatory profile compared to term controls, suggestive of a complex neural environment with concurrent damaging and reparative signals. We propose that limitation of pro-inflammatory responses, which occur early after perinatal insult, may prevent expression of myelination-suppressive genes and support healthy white matter development.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA