Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Glia ; 71(3): 524-540, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36334067

RESUMEN

Genomic analyses have revealed heterogeneity among glial progenitor cells (GPCs), but the compartment selectivity of human GPCs (hGPCs) is unclear. Here, we asked if GPCs of human grey and white brain matter are distinct in their architecture and associated gene expression. RNA profiling of NG2-defined hGPCs derived from adult human neocortex and white matter differed in their expression of genes involved in Wnt, NOTCH, BMP and TGFß signaling, suggesting compartment-selective biases in fate and self-renewal. White matter hGPCs over-expressed the BMP antagonists BAMBI and CHRDL1, suggesting their tonic suppression of astrocytic fate relative to cortical hGPCs, whose relative enrichment of cytoskeletal genes presaged their greater morphological complexity. In human glial chimeric mice, cortical hGPCs assumed larger and more complex morphologies than white matter hGPCs, and both were more complex than their mouse counterparts. These findings suggest that human grey and white matter GPCs comprise context-specific pools with distinct functional biases.


Asunto(s)
Sustancia Gris , Sustancia Blanca , Humanos , Adulto , Animales , Ratones , Sustancia Gris/metabolismo , Neuroglía/metabolismo , Células Madre/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Sustancia Blanca/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Ojo/metabolismo , Proteínas del Tejido Nervioso/metabolismo
2.
J Neurosci ; 34(48): 16153-61, 2014 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-25429155

RESUMEN

Neonatally transplanted human glial progenitor cells (hGPCs) densely engraft and myelinate the hypomyelinated shiverer mouse. We found that, in hGPC-xenografted mice, the human donor cells continue to expand throughout the forebrain, systematically replacing the host murine glia. The differentiation of the donor cells is influenced by the host environment, such that more donor cells differentiated as oligodendrocytes in the hypomyelinated shiverer brain than in myelin wild-types, in which hGPCs were more likely to remain as progenitors. Yet in each recipient, both the number and relative proportion of mouse GPCs fell as a function of time, concomitant with the mitotic expansion and spread of donor hGPCs. By a year after neonatal xenograft, the forebrain GPC populations of implanted mice were largely, and often entirely, of human origin. Thus, neonatally implanted hGPCs outcompeted and ultimately replaced the host population of mouse GPCs, ultimately generating mice with a humanized glial progenitor population. These human glial chimeric mice should permit us to define the specific contributions of glia to a broad variety of neurological disorders, using human cells in vivo.


Asunto(s)
Quimera/fisiología , Células Madre Fetales/fisiología , Células Madre Fetales/trasplante , Neuroglía/fisiología , Neuroglía/trasplante , Prosencéfalo/fisiología , Animales , Animales Recién Nacidos , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Prosencéfalo/citología , Trasplante de Células Madre/métodos
3.
Nat Biotechnol ; 2023 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-37460676

RESUMEN

Competition among adult brain cells has not been extensively researched. To investigate whether healthy glia can outcompete diseased human glia in the adult forebrain, we engrafted wild-type (WT) human glial progenitor cells (hGPCs) produced from human embryonic stem cells into the striata of adult mice that had been neonatally chimerized with mutant Huntingtin (mHTT)-expressing hGPCs. The WT hGPCs outcompeted and ultimately eliminated their human Huntington's disease (HD) counterparts, repopulating the host striata with healthy glia. Single-cell RNA sequencing revealed that WT hGPCs acquired a YAP1/MYC/E2F-defined dominant competitor phenotype upon interaction with the host HD glia. WT hGPCs also outcompeted older resident isogenic WT cells that had been transplanted neonatally, suggesting that competitive success depended primarily on the relative ages of competing populations, rather than on the presence of mHTT. These data indicate that aged and diseased human glia may be broadly replaced in adult brain by younger healthy hGPCs, suggesting a therapeutic strategy for the replacement of aged and diseased human glia.

4.
Hum Mol Genet ; 17(R1): R76-83, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18632701

RESUMEN

The pediatric leukodystrophies comprise a category of disease manifested by neonatal or childhood deficiencies in myelin production or maintenance; these may be due to hereditary defects in one or more genes critical to the initiation of myelination, as in Pelizaeus-Merzbacher Disease, or to enzymatic deficiencies with aberrant substrate accumulation-related dysfunction, as in the lysosomal storage disorders. Despite differences in both phenotype and natural history, these disorders are all essentially manifested by a profound deterioration in neurological function with age. A congenital deficit in forebrain myelination is also noted in children with the periventricular leukomalacia of cerebral palsy, another major source of neurological morbidity. In light of the wide range of disorders to which congenital hypomyelination and/or postnatal demyelination may contribute, and the relative homogeneity of central oligodendrocytes and their progenitors, the pediatric leukodystrophies may be especially attractive targets for cell-based therapeutic strategies. As a result, glial progenitor cells (GPCs), which can give rise to new myelinogenic oligodendrocytes, have become of great interest as potential therapeutic vectors for the restoration of myelin to the hypomyelinated or dysmyelinated childhood CNS. In addition, by distributing themselves throughout the deficient host neuraxis after perinatal allograft, and giving rise to astrocytes as well as oligodendrocytes, glial progenitors appear to be of potential great utility in rectifying enzymatic deficiencies. In this review, we focus on current efforts to develop the use of isolated human GPCs as transplantable agents both for mediating enzymatic restoration to the enzyme-deficient brain and for therapeutic myelination in the disorders of congenital hypomyelination.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Enfermedades Desmielinizantes/terapia , Vaina de Mielina/metabolismo , Trasplante de Células Madre , Niño , Enfermedades Desmielinizantes/congénito , Enfermedades Desmielinizantes/inmunología , Humanos , Leucodistrofia de Células Globoides/inmunología , Leucodistrofia de Células Globoides/terapia , Enfermedades por Almacenamiento Lisosomal/inmunología , Enfermedades por Almacenamiento Lisosomal/terapia , Neuroglía/inmunología , Neuroglía/trasplante , Células Madre/inmunología
5.
Cell Rep ; 31(7): 107658, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32433967

RESUMEN

Neonatally transplanted human glial progenitor cells (hGPCs) can myelinate the brains of myelin-deficient shiverer mice, rescuing their phenotype and survival. Yet, it has been unclear whether implanted hGPCs are similarly able to remyelinate the diffusely demyelinated adult CNS. We, therefore, ask if hGPCs could remyelinate both congenitally hypomyelinated adult shiverers and normal adult mice after cuprizone demyelination. In adult shiverers, hGPCs broadly disperse and differentiate as myelinating oligodendrocytes after subcortical injection, improving both host callosal conduction and ambulation. Implanted hGPCs similarly remyelinate denuded axons after cuprizone demyelination, whether delivered before or after demyelination. RNA sequencing (RNA-seq) of hGPCs back from cuprizone-demyelinated brains reveals their transcriptional activation of oligodendrocyte differentiation programs, while distinguishing them from hGPCs not previously exposed to demyelination. These data indicate the ability of transplanted hGPCs to disperse throughout the adult CNS, to broadly myelinate regions of dysmyelination, and also to be recruited as myelinogenic oligodendrocytes later in life, upon demyelination-associated demand.


Asunto(s)
Encéfalo/fisiopatología , Enfermedades Desmielinizantes/genética , Neuroglía/metabolismo , Células Madre/metabolismo , Animales , Diferenciación Celular , Humanos , Ratones
6.
Cell Stem Cell ; 21(2): 195-208.e6, 2017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-28736215

RESUMEN

In this study, we investigated whether intrinsic glial dysfunction contributes to the pathogenesis of schizophrenia (SCZ). Our approach was to establish humanized glial chimeric mice using glial progenitor cells (GPCs) produced from induced pluripotent stem cells derived from patients with childhood-onset SCZ. After neonatal implantation into myelin-deficient shiverer mice, SCZ GPCs showed premature migration into the cortex, leading to reduced white matter expansion and hypomyelination relative to controls. The SCZ glial chimeras also showed delayed astrocytic differentiation and abnormal astrocytic morphologies. When established in myelin wild-type hosts, SCZ glial mice showed reduced prepulse inhibition and abnormal behavior, including excessive anxiety, antisocial traits, and disturbed sleep. RNA-seq of cultured SCZ human glial progenitor cells (hGPCs) revealed disrupted glial differentiation-associated and synaptic gene expression, indicating that glial pathology was cell autonomous. Our data therefore suggest a causal role for impaired glial maturation in the development of schizophrenia and provide a humanized model for its in vivo assessment.


Asunto(s)
Quimera/metabolismo , Células Madre Pluripotentes Inducidas/patología , Neuroglía/patología , Esquizofrenia/patología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Conducta , Diferenciación Celular/genética , Regulación de la Expresión Génica , Humanos , Ratones , Vaina de Mielina/metabolismo , Neuroglía/metabolismo , Fenotipo , Esquizofrenia/genética
7.
J Clin Invest ; 124(12): 5323-36, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25401469

RESUMEN

Progressive multifocal leukoencephalopathy (PML) is a demyelinating disease triggered by infection with the human gliotropic JC virus (JCV). Due to the human-selective nature of the virus, there are no animal models available to investigate JCV pathogenesis. To address this issue, we developed mice with humanized white matter by engrafting human glial progenitor cells (GPCs) into neonatal immunodeficient and myelin-deficient mice. Intracerebral delivery of JCV resulted in infection and subsequent demyelination of these chimeric mice. Human GPCs and astrocytes were infected more readily than oligodendrocytes, and viral replication was noted primarily in human astrocytes and GPCs rather than oligodendrocytes, which instead expressed early viral T antigens and exhibited apoptotic death. Engraftment of human GPCs in normally myelinated and immunodeficient mice resulted in humanized white matter that was chimeric for human astrocytes and GPCs. JCV effectively propagated in these mice, which indicates that astroglial infection is sufficient for JCV spread. Sequencing revealed progressive mutation of the JCV capsid protein VP1 after infection, suggesting that PML may evolve with active infection. These results indicate that the principal CNS targets for JCV infection are astrocytes and GPCs and that infection is associated with progressive mutation, while demyelination is a secondary occurrence, following T antigen-triggered oligodendroglial apoptosis. More broadly, this study provides a model by which to further assess the biology and treatment of human-specific gliotropic viruses.


Asunto(s)
Astrocitos/inmunología , Virus JC/fisiología , Leucoencefalopatía Multifocal Progresiva/inmunología , Trasplante de Células Madre , Células Madre/inmunología , Quimera por Trasplante/inmunología , Replicación Viral/inmunología , Animales , Antígenos Virales de Tumores/genética , Antígenos Virales de Tumores/inmunología , Apoptosis/genética , Apoptosis/inmunología , Astrocitos/patología , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Modelos Animales de Enfermedad , Femenino , Xenoinjertos , Humanos , Leucoencefalopatía Multifocal Progresiva/genética , Leucoencefalopatía Multifocal Progresiva/patología , Masculino , Ratones , Células Madre/patología
8.
Cell Stem Cell ; 12(2): 252-64, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23395447

RESUMEN

Neonatal engraftment by oligodendrocyte progenitor cells (OPCs) permits the myelination of the congenitally dysmyelinated brain. To establish a potential autologous source of these cells, we developed a strategy by which to differentiate human induced pluripotent stem cells (hiPSCs) into OPCs. From three hiPSC lines, as well as from human embryonic stem cells (hESCs), we generated highly enriched OLIG2(+)/PDGFRα(+)/NKX2.2(+)/SOX10(+) human OPCs, which could be further purified using fluorescence-activated cell sorting. hiPSC OPCs efficiently differentiated into both myelinogenic oligodendrocytes and astrocytes, in vitro and in vivo. Neonatally engrafted hiPSC OPCs robustly myelinated the brains of myelin-deficient shiverer mice and substantially increased their survival. The speed and efficiency of myelination by hiPSC OPCs was higher than that previously observed using fetal-tissue-derived OPCs, and no tumors from these grafts were noted as long as 9 months after transplant. These results suggest the potential utility of hiPSC-derived OPCs in treating disorders of myelin loss.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/terapia , Células Madre Pluripotentes Inducidas/citología , Vaina de Mielina/metabolismo , Oligodendroglía/citología , Células Madre/citología , Células Madre/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Línea Celular , Células Cultivadas , Citometría de Flujo , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio , Humanos , Inmunohistoquímica , Ratones , Proteínas Nucleares , Factores de Transcripción
9.
Nat Biotechnol ; 29(10): 934-41, 2011 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-21947029

RESUMEN

Experimental animals with myelin disorders can be treated by transplanting oligodendrocyte progenitor cells (OPCs) into the affected brain or spinal cord. OPCs have been isolated by their expression of gangliosides recognized by mAb A2B5, but this marker also identifies lineage-restricted astrocytes and immature neurons. To establish a more efficient means of isolating myelinogenic OPCs, we sorted fetal human forebrain cells for CD140a, an epitope of platelet derived growth factor receptor (PDGFR)α, which is differentially expressed by OPCs. CD140a(+) cells were isolated as mitotic bipotential progenitors that initially expressed neither mature neuronal nor astrocytic phenotypic markers, yet could be instructed to either oligodendrocyte or astrocyte fate in vitro. Transplanted CD140a(+) cells were highly migratory and robustly myelinated the hypomyelinated shiverer mouse brain more rapidly and efficiently than did A2B5(+)cells. Microarray analysis of CD140a(+) cells revealed overexpression of the oligodendroglial marker CD9, suggesting that CD9(+)/CD140a(+) cells may constitute an even more highly enriched population of myelinogenic progenitor cells.


Asunto(s)
Movimiento Celular , Vaina de Mielina/metabolismo , Oligodendroglía/citología , Oligodendroglía/trasplante , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Trasplante de Células Madre , Células Madre/citología , Animales , Astrocitos/citología , Astrocitos/metabolismo , Axones/metabolismo , Proliferación Celular , Corteza Cerebral/citología , Corteza Cerebral/embriología , Células Madre Fetales/citología , Células Madre Fetales/metabolismo , Feto/citología , Regulación de la Expresión Génica , Humanos , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/genética , Células Madre/metabolismo , Tetraspanina 29/metabolismo
10.
Cell Stem Cell ; 2(6): 553-65, 2008 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-18522848

RESUMEN

Congenitally hypomyelinated shiverer mice fail to generate compact myelin and die by 18-21 weeks of age. Using multifocal anterior and posterior fossa delivery of sorted fetal human glial progenitor cells into neonatal shiverer x rag2(-/-) mice, we achieved whole neuraxis myelination of the engrafted hosts, which in a significant fraction of cases rescued this otherwise lethal phenotype. The transplanted mice exhibited greatly prolonged survival with progressive resolution of their neurological deficits. Substantial myelination in multiple regions was accompanied by the acquisition of normal nodes of Ranvier and transcallosal conduction velocities, ultrastructurally normal and complete myelination of most axons, and a restoration of a substantially normal neurological phenotype. Notably, the resultant mice were cerebral chimeras, with murine gray matter but a predominantly human white matter glial composition. These data demonstrate that the neonatal transplantation of human glial progenitor cells can effectively treat disorders of congenital and perinatal hypomyelination.


Asunto(s)
Células Madre Adultas/trasplante , Cuerpo Calloso/trasplante , Vaina de Mielina/metabolismo , Vaina de Mielina/trasplante , Neuroglía/trasplante , Trasplante de Células Madre , Células Madre Adultas/metabolismo , Agenesia del Cuerpo Calloso , Animales , Animales Recién Nacidos/anomalías , Animales Recién Nacidos/embriología , Tratamiento Basado en Trasplante de Células y Tejidos , Cuerpo Calloso/embriología , Cuerpo Calloso/metabolismo , Enfermedades Desmielinizantes/congénito , Enfermedades Desmielinizantes/terapia , Humanos , Huésped Inmunocomprometido , Ratones , Vaina de Mielina/genética , Conducción Nerviosa , Neuroglía/metabolismo , Nódulos de Ranvier/metabolismo , Nódulos de Ranvier/trasplante , Distribución Tisular , Quimera por Trasplante/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA