Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Brain ; 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39133566

RESUMEN

Progressive multifocal leukoencephalopathy (PML) is a demyelinating infection of the immunosuppressed brain, mediated by the gliotropic polyomavirus JCV. JCV replicates in human glial progenitor cells and astrocytes, which undergo viral T antigen-triggered mitosis, enabling viral replication. We asked if JCV spread might therefore be accelerated by glial proliferation. Using both in vitro analysis and a human glial chimeric mouse model of JCV infection, we found that dividing human astrocytes supported JCV propagation to a substantially greater degree than did mitotically quiescent cells. Accordingly, bulk and single cell RNA-sequence analysis revealed that JCV-infected glia differentially manifested cell cycle-linked disruption of both DNA damage response and transcriptional regulatory pathways. In vivo, JCV infection of humanized glial chimeras was greatly accentuated by cuprizone-induced demyelination and its associated mobilization of GPCs. Importantly, in vivo infection triggered the death of uninfected as well as infected glia, reflecting significant bystander death. Together, these data suggest that JCV propagation in PML may be accelerated by glial cell division. As such, the accentuated glial proliferation attending disease-associated demyelination may provide an especially favorable environment for JCV propagation, thus potentiating oligodendrocytic bystander death and further accelerating demyelination in susceptible hosts.

2.
Am J Hum Genet ; 100(4): 617-634, 2017 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-28366443

RESUMEN

Pelizaeus-Merzbacher disease (PMD) is a pediatric disease of myelin in the central nervous system and manifests with a wide spectrum of clinical severities. Although PMD is a rare monogenic disease, hundreds of mutations in the X-linked myelin gene proteolipid protein 1 (PLP1) have been identified in humans. Attempts to identify a common pathogenic process underlying PMD have been complicated by an incomplete understanding of PLP1 dysfunction and limited access to primary human oligodendrocytes. To address this, we generated panels of human induced pluripotent stem cells (hiPSCs) and hiPSC-derived oligodendrocytes from 12 individuals with mutations spanning the genetic and clinical diversity of PMD-including point mutations and duplication, triplication, and deletion of PLP1-and developed an in vitro platform for molecular and cellular characterization of all 12 mutations simultaneously. We identified individual and shared defects in PLP1 mRNA expression and splicing, oligodendrocyte progenitor development, and oligodendrocyte morphology and capacity for myelination. These observations enabled classification of PMD subgroups by cell-intrinsic phenotypes and identified a subset of mutations for targeted testing of small-molecule modulators of the endoplasmic reticulum stress response, which improved both morphologic and myelination defects. Collectively, these data provide insights into the pathogeneses of a variety of PLP1 mutations and suggest that disparate etiologies of PMD could require specific treatment approaches for subsets of individuals. More broadly, this study demonstrates the versatility of a hiPSC-based panel spanning the mutational heterogeneity within a single disease and establishes a widely applicable platform for genotype-phenotype correlation and drug screening in any human myelin disorder.


Asunto(s)
Oligodendroglía/patología , Enfermedad de Pelizaeus-Merzbacher/genética , Enfermedad de Pelizaeus-Merzbacher/patología , Técnicas de Cultivo de Célula , Niño , Preescolar , Estrés del Retículo Endoplásmico , Femenino , Humanos , Células Madre Pluripotentes Inducidas/patología , Masculino , Proteína Proteolipídica de la Mielina , Oligodendroglía/metabolismo
3.
Stem Cells ; 35(2): 311-315, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27882623

RESUMEN

Pelizaeus-Merzbacher disease (PMD) is an X-linked disorder caused by mutation in the proteolipid protein-1 (PLP1) gene, which encodes the proteolipid protein of myelinating oligodendroglia. PMD exhibits phenotypic variability that reflects its considerable genotypic heterogeneity, but all forms of the disease result in central hypomyelination, associated in most cases with early neurological dysfunction, progressive deterioration, and ultimately death. PMD may present as a connatal, classic and transitional forms, or as the less severe spastic paraplegia type 2 and PLP-null phenotypes. These disorders are most often associated with duplications of the PLP1 gene, but can also be caused by coding and noncoding point mutations as well as full or partial deletion of the gene. A number of genetically-distinct but phenotypically-similar disorders of hypomyelination exist which, like PMD, lack any effective therapy. Yet as relatively pure CNS hypomyelinating disorders, with limited involvement of the PNS and relatively little attendant neuronal pathology, PMD and similar hypomyelinating disorders are attractive therapeutic targets for neural stem cell and glial progenitor cell transplantation, efforts at which are now underway in a number of research centers. Stem Cells 2017;35:311-315.


Asunto(s)
Enfermedad de Pelizaeus-Merzbacher/terapia , Trasplante de Células Madre , Células Madre/citología , Animales , Modelos Animales de Enfermedad , Humanos , Mutación/genética , Vaina de Mielina/metabolismo , Enfermedad de Pelizaeus-Merzbacher/patología
4.
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
5.
Glia ; 63(8): 1483-93, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26010831

RESUMEN

As new methods for producing and isolating human glial progenitor cells (hGPCs) have been developed, the disorders of myelin have become especially compelling targets for cell-based therapy. Yet as animal modeling of glial progenitor cell-based therapies has progressed, it has become clear that transplanted hGPCs not only engraft and expand within murine hosts, but dynamically outcompete the resident progenitors so as to ultimately dominate the host brain. The engrafted human progenitor cells proceed to generate parenchymal astrocytes, and when faced with a hypomyelinated environment, oligodendrocytes as well. As a result, the recipient brains may become inexorably humanized with regards to their resident glial populations, yielding human glial chimeric mouse brains. These brains provide us a fundamentally new tool by which to assess the species-specific attributes of glia in modulating human cognition and information processing. In addition, the cellular humanization of these brains permits their use in studying glial infectious and inflammatory disorders unique to humans, and the effects of those disorders on the glial contributions to cognition. Perhaps most intriguingly, by pairing our ability to construct human glial chimeras with the production of patient-specific hGPCs derived from pluripotential stem cells, we may now establish mice in which a substantial proportion of resident glia are both human and disease-derived. These mice in particular may provide us new opportunities for studying the human-specific contributions of glia to psychopathology, as well as to higher cognition. As such, the assessment of human glial chimeric mice may provide us new insight into the species-specific contributions of glia to human cognitive evolution, as well as to the pathogenesis of human neurological and neuropsychiatric disease.


Asunto(s)
Enfermedades del Sistema Nervioso Central/fisiopatología , Cognición/fisiología , Células-Madre Neurales/fisiología , Células-Madre Neurales/trasplante , Neuroglía/citología , Neuroglía/fisiología , Animales , Enfermedades del Sistema Nervioso Central/cirugía , Quimera , Humanos , Ratones , Modelos Animales
6.
Cell Rep ; 42(9): 113130, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37708026

RESUMEN

The naked mole rat (NMR) is the longest-lived rodent, resistant to multiple age-related diseases including neurodegeneration. However, the mechanisms underlying the NMR's resistance to neurodegenerative diseases remain elusive. Here, we isolated oligodendrocyte progenitor cells (OPCs) from NMRs and compared their transcriptome with that of other mammals. Extracellular matrix (ECM) genes best distinguish OPCs of long- and short-lived species. Notably, expression levels of CD44, an ECM-binding protein that has been suggested to contribute to NMR longevity by mediating the effect of hyaluronan (HA), are not only high in OPCs of long-lived species but also positively correlate with longevity in multiple cell types/tissues. We found that CD44 localizes to the endoplasmic reticulum (ER) and enhances basal ATF6 activity. CD44 modifies proteome and membrane properties of the ER and enhances ER stress resistance in a manner dependent on unfolded protein response regulators without the requirement of HA. HA-independent role of CD44 in proteostasis regulation may contribute to mammalian longevity.


Asunto(s)
Estrés del Retículo Endoplásmico , Longevidad , Animales , Longevidad/fisiología , Respuesta de Proteína Desplegada , Transcriptoma , Ratas Topo
7.
Nat Med ; 10(1): 93-7, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14702638

RESUMEN

Both late-gestation and adult human forebrain contain large numbers of oligodendrocyte progenitor cells (OPCs). These cells may be identified by their A2B5(+)PSA-NCAM(-) phenotype (positive for the early oligodendrocyte marker A2B5 and negative for the polysialylated neural cell adhesion molecule). We used dual-color fluorescence-activated cell sorting (FACS) to extract OPCs from 21- to 23-week-old fetal human forebrain, and A2B5 selection to extract these cells from adult white matter. When xenografted to the forebrains of newborn shiverer mice, fetal OPCs dispersed throughout the white matter and developed into oligodendrocytes and astrocytes. By 12 weeks, the host brains showed extensive myelin production, compaction and axonal myelination. Isolates of OPCs derived from adult human white matter also myelinated shiverer mouse brain, but much more rapidly than their fetal counterparts, achieving widespread and dense myelin basic protein (MBP) expression by 4 weeks after grafting. Adult OPCs generated oligodendrocytes more efficiently than fetal OPCs, and ensheathed more host axons per donor cell than fetal cells. Both fetal and adult OPC phenotypes mediated the extensive and robust myelination of congenitally dysmyelinated host brain, although their differences suggested their use for different disease targets.


Asunto(s)
Enfermedades Desmielinizantes/terapia , Feto/citología , Proteína Básica de Mielina/biosíntesis , Oligodendroglía/citología , Células Madre/citología , Adulto , Animales , Enfermedades Desmielinizantes/patología , Humanos , Inmunohistoquímica , Ratones , Microscopía Confocal , Microscopía Electrónica
8.
Cell Rep ; 36(1): 109308, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34233199

RESUMEN

Glial pathology is a causal contributor to the striatal neuronal dysfunction of Huntington's disease (HD). We investigate mutant HTT-associated changes in gene expression by mouse and human striatal astrocytes, as well as in mouse microglia, to identify commonalities in glial pathobiology across species and models. Mouse striatal astrocytes are fluorescence-activated cell sorted (FACS) from R6/2 and zQ175 mice, which respectively express exon1-only or full-length mHTT, and human astrocytes are generated either from human embryonic stem cells (hESCs) expressing full-length mHTT or from fetal striatal astrocytes transduced with exon1-only mHTT. Comparison of differential gene expression across these conditions, all with respect to normal HTT controls, reveals cell-type-specific changes in transcription common to both species, yet with differences that distinguish glia expressing truncated mHTT versus full-length mHTT. These data indicate that the differential gene expression of glia expressing truncated mHTT may differ from that of cells expressing full-length mHTT, while identifying a conserved set of dysregulated pathways in HD glia.


Asunto(s)
Enfermedad de Huntington/patología , Neuroglía/patología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Vías Biosintéticas , Colesterol/biosíntesis , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Ratones Endogámicos C57BL , Proteínas Mutantes/metabolismo , Neuroglía/metabolismo , Transcripción Genética
9.
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
10.
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
11.
Cell Rep ; 27(13): 3832-3843.e6, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31242417

RESUMEN

Astrocytic differentiation is developmentally impaired in patients with childhood-onset schizophrenia (SCZ). To determine why, we used genetic gain- and loss-of-function studies to establish the contributions of differentially expressed transcriptional regulators to the defective differentiation of glial progenitor cells (GPCs) produced from SCZ patient-derived induced pluripotent cells (iPSCs). Negative regulators of the bone morphogenetic protein (BMP) pathway were upregulated in SCZ GPCs, including BAMBI, FST, and GREM1, whose overexpression retained SCZ GPCs at the progenitor stage. SMAD4 knockdown (KD) suppressed the production of these BMP inhibitors by SCZ GPCs and rescued normal astrocytic differentiation. In addition, the BMP-regulated transcriptional repressor REST was upregulated in SCZ GPCs, and its KD similarly restored normal glial differentiation. REST KD also rescued potassium-transport-associated gene expression and K+ uptake, which were otherwise deficient in SCZ glia. These data suggest that the glial differentiation defect in childhood-onset SCZ, and its attendant disruption in K+ homeostasis, may be rescued by targeting BMP/SMAD4- and REST-dependent transcription.


Asunto(s)
Diferenciación Celular , Neuroglía/metabolismo , Proteínas Represoras/metabolismo , Esquizofrenia/metabolismo , Transducción de Señal , Proteína Smad4/metabolismo , Adolescente , Adulto , Línea Celular , Niño , Femenino , Humanos , Masculino , Neuroglía/patología , Proteínas Represoras/genética , Esquizofrenia/genética , Esquizofrenia/patología , Proteína Smad4/genética
12.
Cell Stem Cell ; 24(1): 107-122.e7, 2019 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-30554964

RESUMEN

Huntington's disease (HD) is characterized by hypomyelination and neuronal loss. To assess the basis for myelin loss in HD, we generated bipotential glial progenitor cells (GPCs) from human embryonic stem cells (hESCs) derived from mutant Huntingtin (mHTT) embryos or normal controls and performed RNA sequencing (RNA-seq) to assess mHTT-dependent changes in gene expression. In human GPCs (hGPCs) derived from 3 mHTT hESC lines, transcription factors associated with glial differentiation and myelin synthesis were sharply downregulated relative to normal hESC GPCs; NKX2.2, OLIG2, SOX10, MYRF, and their downstream targets were all suppressed. Accordingly, when mHTT hGPCs were transplanted into hypomyelinated shiverer mice, the resultant glial chimeras were hypomyelinated; this defect could be rescued by forced expression of SOX10 and MYRF by mHTT hGPCs. The mHTT hGPCs also manifested impaired astrocytic differentiation and developed abnormal fiber architecture. White matter involution in HD is thus a product of the cell-autonomous, mHTT-dependent suppression of glial differentiation.


Asunto(s)
Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Células Madre Embrionarias Humanas/patología , Proteína Huntingtina/genética , Enfermedad de Huntington/patología , Neuroglía/patología , Células Madre/patología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Diferenciación Celular , Quimera , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/metabolismo , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio , Células Madre Embrionarias Humanas/metabolismo , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Ratones , Mutación , Neurogénesis , Neuroglía/metabolismo , Proteínas Nucleares , Células Madre/metabolismo , Factores de Transcripción
13.
Nat Biotechnol ; 22(3): 297-305, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14990951

RESUMEN

Lineage-restricted progenitors of the central nervous system (CNS) are not readily expandable because their mitotic competence is limited. Here we used retroviral overexpression of human telomerase reverse transcriptase (hTERT) to immortalize progenitors from human fetal spinal cord. The hTERT-immortalized cells divided in basic fibroblast growth factor (bFGF) expressed high telomerase activity, and gave rise to phenotypically restricted subpopulations of either glia or neurons. The latter included a prototypic line, hSC11V-TERT, that gave rise only to neurons. These included both chx10(+) interneurons and Islet1(+)/Hb9(+)/ChAT(+) motor neurons; the latter were recognized by green fluorescent protein (GFP) driven by the Hb9 enhancer. The neurons were postmitotic and achieved electrophysiologic competence. Upon xenograft to both fetal rat brain and injured adult spinal cord, they matured as neurons and survived for 6 months, with no evident tumorigenesis. The cells have survived >168 doublings in vitro, with karyotypic normalcy and without replicative senescence. hTERT overexpression thus permits the generation of progenitor lines able to give rise to phenotypically restricted neurons.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Neuronas/citología , Neuronas/fisiología , Células Madre/citología , Células Madre/fisiología , Telomerasa/metabolismo , Ingeniería de Tejidos/métodos , Diferenciación Celular/fisiología , División Celular/fisiología , Línea Celular , Supervivencia Celular/fisiología , Proteínas de Unión al ADN , Mejoramiento Genético/métodos , Humanos , Enfermedades del Sistema Nervioso/cirugía , Plasticidad Neuronal/fisiología , Proteínas de los Retroviridae/genética , Proteínas de los Retroviridae/metabolismo , Médula Espinal/citología , Médula Espinal/embriología , Médula Espinal/fisiología , Trasplante de Células Madre/métodos , Telomerasa/genética
14.
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
15.
Nat Commun ; 7: 11758, 2016 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-27273432

RESUMEN

The causal contribution of glial pathology to Huntington disease (HD) has not been heavily explored. To define the contribution of glia to HD, we established human HD glial chimeras by neonatally engrafting immunodeficient mice with mutant huntingtin (mHTT)-expressing human glial progenitor cells (hGPCs), derived from either human embryonic stem cells or mHTT-transduced fetal hGPCs. Here we show that mHTT glia can impart disease phenotype to normal mice, since mice engrafted intrastriatally with mHTT hGPCs exhibit worse motor performance than controls, and striatal neurons in mHTT glial chimeras are hyperexcitable. Conversely, normal glia can ameliorate disease phenotype in transgenic HD mice, as striatal transplantation of normal glia rescues aspects of electrophysiological and behavioural phenotype, restores interstitial potassium homeostasis, slows disease progression and extends survival in R6/2 HD mice. These observations suggest a causal role for glia in HD, and further suggest a cell-based strategy for disease amelioration in this disorder.


Asunto(s)
Enfermedad de Huntington/patología , Neuroglía/patología , Animales , Conducta Animal , Quimera/metabolismo , Cognición , Cruzamientos Genéticos , Progresión de la Enfermedad , Femenino , Células Madre Embrionarias Humanas/metabolismo , Humanos , Proteína Huntingtina/metabolismo , Receptores de Hialuranos/metabolismo , Masculino , Ratones , Actividad Motora , Neostriado/patología , Neuroglía/metabolismo , Neuronas/metabolismo , Fenotipo , Trasplante de Células Madre , Análisis de Supervivencia
16.
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
17.
Cell Stem Cell ; 12(3): 342-53, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23472873

RESUMEN

Human astrocytes are larger and more complex than those of infraprimate mammals, suggesting that their role in neural processing has expanded with evolution. To assess the cell-autonomous and species-selective properties of human glia, we engrafted human glial progenitor cells (GPCs) into neonatal immunodeficient mice. Upon maturation, the recipient brains exhibited large numbers and high proportions of both human glial progenitors and astrocytes. The engrafted human glia were gap-junction-coupled to host astroglia, yet retained the size and pleomorphism of hominid astroglia, and propagated Ca2+ signals 3-fold faster than their hosts. Long-term potentiation (LTP) was sharply enhanced in the human glial chimeric mice, as was their learning, as assessed by Barnes maze navigation, object-location memory, and both contextual and tone fear conditioning. Mice allografted with murine GPCs showed no enhancement of either LTP or learning. These findings indicate that human glia differentially enhance both activity-dependent plasticity and learning in mice.


Asunto(s)
Aprendizaje/fisiología , Neuroglía/citología , Plasticidad Neuronal/fisiología , Prosencéfalo/citología , Células Madre/citología , Sinapsis/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Encéfalo , Calcio/metabolismo , Diferenciación Celular , Hipocampo/citología , Humanos , Ratones , Neuroglía/metabolismo , Prosencéfalo/metabolismo , Células Madre/metabolismo
18.
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
19.
Science ; 338(6106): 491-5, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-23112326

RESUMEN

The diseases of myelin are among the most prevalent and disabling conditions in neurology. These diseases include both the vascular and inflammatory demyelinating disorders of adulthood, as well as the childhood leukodystrophies and cerebral palsy. These fundamentally glial disorders may be amenable to treatment by glial progenitor cells (GPCs), which give rise to astroglia and myelin-producing oligodendrocytes. Given the development of new methods for generating and isolating human GPCs, the myelin disorders may now be compelling targets for cell-based therapy. In addition, the efficient engraftment and expansion of human GPCs in murine hosts has led to the development of human glial chimeric mouse brains, which provides new opportunities for studying the species-specific roles of human glia in cognition, as well as in disease pathogenesis.


Asunto(s)
Enfermedades Desmielinizantes/terapia , Modelos Neurológicos , Células-Madre Neurales/trasplante , Oligodendroglía/trasplante , Adulto , Animales , Niño , Quimera , Modelos Animales de Enfermedad , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Humanos , Ratones , Vaina de Mielina/metabolismo , Células-Madre Neurales/fisiología , Oligodendroglía/metabolismo , Oligodendroglía/fisiología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA