Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 3873, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38719882

RESUMO

Human glial progenitor cells (hGPCs) exhibit diminished expansion competence with age, as well as after recurrent demyelination. Using RNA-sequencing to compare the gene expression of fetal and adult hGPCs, we identify age-related changes in transcription consistent with the repression of genes enabling mitotic expansion, concurrent with the onset of aging-associated transcriptional programs. Adult hGPCs develop a repressive transcription factor network centered on MYC, and regulated by ZNF274, MAX, IKZF3, and E2F6. Individual over-expression of these factors in iPSC-derived hGPCs lead to a loss of proliferative gene expression and an induction of mitotic senescence, replicating the transcriptional changes incurred during glial aging. miRNA profiling identifies the appearance of an adult-selective miRNA signature, imposing further constraints on the expansion competence of aged GPCs. hGPC aging is thus associated with acquisition of a MYC-repressive environment, suggesting that suppression of these repressors of glial expansion may permit the rejuvenation of aged hGPCs.


Assuntos
Envelhecimento , MicroRNAs , Neuroglia , Fatores de Transcrição , Humanos , Neuroglia/metabolismo , Neuroglia/citologia , Envelhecimento/genética , Envelhecimento/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Senescência Celular/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco/metabolismo , Células-Tronco/citologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Adulto , Redes Reguladoras de Genes , Proliferação de Células/genética , Regulação da Expressão Gênica no Desenvolvimento , Perfilação da Expressão Gênica
2.
Cell Rep ; 42(9): 113130, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37708026

RESUMO

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.


Assuntos
Estresse do Retículo Endoplasmático , Longevidade , Animais , Longevidade/fisiologia , Resposta a Proteínas não Dobradas , Transcriptoma , Ratos-Toupeira
3.
Cell Rep ; 40(9): 111291, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-36044851

RESUMO

Huntington's disease (HD) is characterized by defective oligodendroglial differentiation and white matter disease. Here, we investigate the role of oligodendrocyte progenitor cell (OPC) dysfunction in adult myelin maintenance in HD. We first note a progressive, age-related loss of myelin in both R6/2 and zQ175 HD mice compared with wild-type controls. Adult R6/2 mice then manifest a significant delay in remyelination following cuprizone demyelination. RNA-sequencing and proteomic analysis of callosal white matter and OPCs isolated from both R6/2 and zQ175 mice reveals a systematic downregulation of genes associated with oligodendrocyte differentiation and myelinogenesis. Gene co-expression and network analysis predicts repressed Tcf7l2 signaling as a major driver of this expression pattern. In vivo Tcf7l2 overexpression restores both myelin gene expression and remyelination in demyelinated R6/2 mice. These data causally link impaired TCF7L2-dependent transcription to the poor development and homeostatic retention of myelin in HD and provide a mechanism for its therapeutic restoration.


Assuntos
Doenças Desmielinizantes , Doença de Huntington , Remielinização , Animais , Diferenciação Celular/genética , Doenças Desmielinizantes/metabolismo , Doença de Huntington/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Proteômica , Remielinização/fisiologia , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo
4.
Cell Rep ; 36(1): 109308, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34233199

RESUMO

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.


Assuntos
Doença de Huntington/patologia , Neuroglia/patologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Vias Biossintéticas , Colesterol/biossíntese , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Camundongos Endogâmicos C57BL , Proteínas Mutantes/metabolismo , Neuroglia/metabolismo , Transcrição Gênica
5.
Stem Cell Reports ; 15(4): 869-882, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-32976765

RESUMO

Human glial progenitor cells (hGPCs) are promising cellular substrates to explore for the in situ production of new neurons for brain repair. Proof of concept for direct neuronal reprogramming of glial progenitors has been obtained in mouse models in vivo, but conversion using human cells has not yet been demonstrated. Such studies have been difficult to perform since hGPCs are born late during human fetal development, with limited accessibility for in vitro culture. In this study, we show proof of concept of hGPC conversion using fetal cells and also establish a renewable and reproducible stem cell-based hGPC system for direct neural conversion in vitro. Using this system, we have identified optimal combinations of fate determinants for the efficient dopaminergic (DA) conversion of hGPCs, thereby yielding a therapeutically relevant cell type that selectively degenerates in Parkinson's disease. The induced DA neurons show a progressive, subtype-specific phenotypic maturation and acquire functional electrophysiological properties indicative of DA phenotype.


Assuntos
Reprogramação Celular , Neurônios Dopaminérgicos/citologia , Células-Tronco Fetais/citologia , Mesencéfalo/citologia , Células-Tronco Neurais/citologia , Neuroglia/citologia , Neurônios Dopaminérgicos/metabolismo , Células-Tronco Fetais/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Modelos Biológicos , Células-Tronco Neurais/metabolismo , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
6.
Cell Rep ; 31(7): 107658, 2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32433967

RESUMO

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.


Assuntos
Encéfalo/fisiopatologia , Doenças Desmielinizantes/genética , Neuroglia/metabolismo , Células-Tronco/metabolismo , Animais , Diferenciação Celular , Humanos , Camundongos
7.
Cell Rep ; 27(13): 3832-3843.e6, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31242417

RESUMO

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.


Assuntos
Diferenciação Celular , Neuroglia/metabolismo , Proteínas Repressoras/metabolismo , Esquizofrenia/metabolismo , Transdução de Sinais , Proteína Smad4/metabolismo , Adolescente , Adulto , Linhagem Celular , Criança , Feminino , Humanos , Masculino , Neuroglia/patologia , Proteínas Repressoras/genética , Esquizofrenia/genética , Esquizofrenia/patologia , Proteína Smad4/genética
8.
Cell Stem Cell ; 24(1): 107-122.e7, 2019 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-30554964

RESUMO

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.


Assuntos
Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Células-Tronco Embrionárias Humanas/patologia , Proteína Huntingtina/genética , Doença de Huntington/patologia , Neuroglia/patologia , Células-Tronco/patologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Diferenciação Celular , Quimera , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/metabolismo , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Doença de Huntington/genética , Doença de Huntington/metabolismo , Camundongos , Mutação , Neurogênese , Neuroglia/metabolismo , Proteínas Nucleares , Células-Tronco/metabolismo , Fatores de Transcrição
9.
Sci Signal ; 11(515)2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29382785

RESUMO

Fluorescent Ca2+ indicators have been essential for the analysis of Ca2+ signaling events in various cell types. We showed that chemical Ca2+ indicators, but not a genetically encoded Ca2+ indicator, potently suppressed the activity of Na+- and K+-dependent adenosine triphosphatase (Na,K-ATPase), independently of their Ca2+ chelating activity. Loading of commonly used Ca2+ indicators, including Fluo-4 acetoxymethyl (AM), Rhod-2 AM, and Fura-2 AM, and of the Ca2+ chelator BAPTA AM into cultured mouse or human neurons, astrocytes, cardiomyocytes, or kidney proximal tubule epithelial cells suppressed Na,K-ATPase activity by 30 to 80%. Ca2+ indicators also suppressed the agonist-induced activation of the Na,K-ATPase, altered metabolic status, and caused a dose-dependent loss of cell viability. Loading of Ca2+ indicators into mice, which is carried out for two-photon imaging, markedly altered brain extracellular concentrations of K+ and ATP. These results suggest that a critical review of data obtained with chemical Ca2+ indicators may be necessary.


Assuntos
Astrócitos/metabolismo , Inibidores Enzimáticos/farmacologia , Corantes Fluorescentes/farmacologia , Túbulos Renais Proximais/metabolismo , Miócitos Cardíacos/metabolismo , Neurônios/metabolismo , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Compostos de Anilina/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/enzimologia , Células Cultivadas , Fura-2/farmacologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Potássio/metabolismo , Xantenos/farmacologia
10.
Cell Stem Cell ; 21(2): 195-208.e6, 2017 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-28736215

RESUMO

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.


Assuntos
Quimera/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Neuroglia/patologia , Esquizofrenia/patologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Comportamento , Diferenciação Celular/genética , Regulação da Expressão Gênica , Humanos , Camundongos , Bainha de Mielina/metabolismo , Neuroglia/metabolismo , Fenótipo , Esquizofrenia/genética
11.
Nat Commun ; 7: 11758, 2016 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-27273432

RESUMO

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.


Assuntos
Doença de Huntington/patologia , Neuroglia/patologia , Animais , Comportamento Animal , Quimera/metabolismo , Cognição , Cruzamentos Genéticos , Progressão da Doença , Feminino , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Proteína Huntingtina/metabolismo , Receptores de Hialuronatos/metabolismo , Masculino , Camundongos , Atividade Motora , Neostriado/patologia , Neuroglia/metabolismo , Neurônios/metabolismo , Fenótipo , Transplante de Células-Tronco , Análise de Sobrevida
12.
J Clin Invest ; 124(12): 5323-36, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25401469

RESUMO

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.


Assuntos
Astrócitos/imunologia , Vírus JC/fisiologia , Leucoencefalopatia Multifocal Progressiva/imunologia , Transplante de Células-Tronco , Células-Tronco/imunologia , Quimeras de Transplante/imunologia , Replicação Viral/imunologia , Animais , Antígenos Virais de Tumores/genética , Antígenos Virais de Tumores/imunologia , Apoptose/genética , Apoptose/imunologia , Astrócitos/patologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Modelos Animais de Doenças , Feminino , Xenoenxertos , Humanos , Leucoencefalopatia Multifocal Progressiva/genética , Leucoencefalopatia Multifocal Progressiva/patologia , Masculino , Camundongos , Células-Tronco/patologia
13.
J Neurosci ; 34(48): 16153-61, 2014 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-25429155

RESUMO

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.


Assuntos
Quimera/fisiologia , Células-Tronco Fetais/fisiologia , Células-Tronco Fetais/transplante , Neuroglia/fisiologia , Neuroglia/transplante , Prosencéfalo/fisiologia , Animais , Animais Recém-Nascidos , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Prosencéfalo/citologia , Transplante de Células-Tronco/métodos
14.
Cell Rep ; 3(6): 2127-41, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23727239

RESUMO

Glial progenitor cells (GPCs) are a potential source of malignant gliomas. We used A2B5-based sorting to extract tumorigenic GPCs from human gliomas spanning World Health Organization grades II-IV. Messenger RNA profiling identified a cohort of genes that distinguished A2B5+ glioma tumor progenitor cells (TPCs) from A2B5+ GPCs isolated from normal white matter. A core set of genes and pathways was substantially dysregulated in A2B5+ TPCs, which included the transcription factor SIX1 and its principal cofactors, EYA1 and DACH2. Small hairpin RNAi silencing of SIX1 inhibited the expansion of glioma TPCs in vitro and in vivo, suggesting a critical and unrecognized role of the SIX1-EYA1-DACH2 system in glioma genesis or progression. By comparing the expression patterns of glioma TPCs with those of normal GPCs, we have identified a discrete set of pathways by which glial tumorigenesis may be better understood and more specifically targeted.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Glioma/genética , Glioma/patologia , Células-Tronco Neoplásicas/patologia , Neuroglia/patologia , Neuroglia/fisiologia , Adulto , Neoplasias Encefálicas/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Humanos , Pessoa de Meia-Idade , Neuroglia/metabolismo , Ativação Transcricional
15.
Cell Calcium ; 53(4): 297-301, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23462235

RESUMO

Earlier studies have shown that activation of adenosine A1 receptors on peripheral pain fibers contributes to acupuncture-induced suppression of painful input. In addition to adenosine, acupuncture triggers the release of other purines, including ATP and ADP that may bind to purine receptors on nearby fibroblasts. We here show that purine agonists trigger increase in cytosolic Ca(2+) signaling in a cultured human fibroblasts cell line. The profile of agonist-induced Ca(2+) increases indicates that the cells express functional P2yR2 and P2yR4 receptors, as well as P2yR1 and P2xR7 receptors. Unexpectedly, purine-induced Ca(2+) signaling was associated with a remodeling of the actin cytoskeleton. ATP induced a transient loss in F-actin stress fiber. The changes of actin cytoskeleton occurred slowly and peaked at 10min after agonist exposure. Inhibition of ATP-induced increases in Ca(2+) by cyclopiazonic acid blocked receptor-mediated cytoskeleton remodeling. The Ca(2+) ionophore failed to induce cytoskeletal remodeling despite triggering robust increases in cytosolic Ca(2+). These observations indicate that purine signaling induces transient changes in fibroblast cytoarchitecture that could be related to the beneficial effects of acupuncture.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Receptores Purinérgicos P2/metabolismo , Adenosina Trifosfatases/antagonistas & inibidores , Adenosina Trifosfatases/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio , Células Cultivadas , Humanos , Indóis/farmacologia , Receptores Purinérgicos P2/biossíntese , Relação Estrutura-Atividade
16.
Cell Stem Cell ; 12(2): 252-64, 2013 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-23395447

RESUMO

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.


Assuntos
Doença de Charcot-Marie-Tooth/terapia , Células-Tronco Pluripotentes Induzidas/citologia , Bainha de Mielina/metabolismo , Oligodendroglia/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Linhagem Celular , Células Cultivadas , Citometria de Fluxo , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio , Humanos , Imuno-Histoquímica , Camundongos , Proteínas Nucleares , Fatores de Transcrição
17.
Cell ; 146(1): 53-66, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21729780

RESUMO

Malignant gliomas are aggressive brain tumors with limited therapeutic options, and improvements in treatment require a deeper molecular understanding of this disease. As in other cancers, recent studies have identified highly tumorigenic subpopulations within malignant gliomas, known generally as cancer stem cells. Here, we demonstrate that glioma stem cells (GSCs) produce nitric oxide via elevated nitric oxide synthase-2 (NOS2) expression. GSCs depend on NOS2 activity for growth and tumorigenicity, distinguishing them from non-GSCs and normal neural progenitors. Gene expression profiling identified many NOS2-regulated genes, including the cell-cycle inhibitor cell division autoantigen-1 (CDA1). Further, high NOS2 expression correlates with decreased survival in human glioma patients, and NOS2 inhibition slows glioma growth in a murine intracranial model. These data provide insight into how GSCs are mechanistically distinct from their less tumorigenic counterparts and suggest that NOS2 inhibition may be an efficacious approach to treating this devastating disease.


Assuntos
Proliferação de Células , Glioma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Animais , Autoantígenos/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/metabolismo , Óxido Nítrico/metabolismo , Células Tumorais Cultivadas
18.
J Neurosci ; 30(44): 14635-48, 2010 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-21048121

RESUMO

Sox2 is expressed by neural stem and progenitor cells, and a sox2 enhancer identifies these cells in the forebrains of both fetal and adult transgenic mouse reporters. We found that an adenovirus encoding EGFP placed under the regulatory control of a 0.4 kb sox2 core enhancer selectively identified multipotential and self-renewing neural progenitor cells in dissociates of human fetal forebrain. Upon EGFP-based fluorescence-activated cell sorting (FACS), the E/sox2:EGFP(+) isolates were propagable for up to 1 year in vitro, and remained multilineage competent throughout. E/sox2:EGFP(+) cells expressed more telomerase enzymatic activity than matched E/sox2:EGFP-depleted populations, and maintained their telomeric lengths with successive passage. Gene expression analysis of E/sox2:EGFP-sorted neural progenitor cells, normalized to the unsorted forebrain dissociates from which they derived, revealed marked overexpression of genes within the notch and wnt pathways, and identified multiple elements of each pathway that appear selective to human neural progenitors. Sox2 enhancer-based FACS thus permits the prospective identification and direct isolation of a telomerase-active population of neural stem cells from the human fetal forebrain, and the elucidation of both the transcriptome and dominant signaling pathways of these critically important cells.


Assuntos
Células-Tronco Embrionárias/citologia , Elementos Facilitadores Genéticos/genética , Citometria de Fluxo/métodos , Células-Tronco Neurais/citologia , Fatores de Transcrição SOXB1/genética , Telomerase/biossíntese , Linhagem da Célula/genética , Separação Celular/métodos , Células Cultivadas , Células-Tronco Embrionárias/classificação , Células-Tronco Embrionárias/enzimologia , Feto , Humanos , Células-Tronco Neurais/classificação , Células-Tronco Neurais/enzimologia , Estudos Prospectivos , Telomerase/genética
19.
Nat Protoc ; 2(11): 2815-25, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18007617

RESUMO

Traditional methods of generating immortalized lines of both somatic cells and their progenitors have relied on the use of oncogenes. However, the resulting lines are typically anaplastic in vitro and tumorigenic in vivo, and hence of limited utility. The overexpression of telomerase, as mediated by the induced overexpression of human telomerase reverse transcriptase (hTERT), has permitted the generation of stable, non-oncogenic lines of a variety of cell types. This strategy for immortalization has found special utility in the central nervous system, as few stable lines are available for the study of either human neural progenitor cells, or of neurons or glia of restricted phenotype. We describe the use of retroviral hTERT overexpression for generating lines of immortalized human neural progenitor cells, whose neuronal progeny are phenotypically restricted, post-mitotic and functionally competent. Although we focus here on telomerase immortalization of spinal neural progenitors, this is a broadly applicable protocol for using hTERT to immortalize human fetal neural progenitors of any pre-selected phenotype and for characterizing the cell lines thereby generated.


Assuntos
Técnicas de Cultura de Células , Células-Tronco Fetais/citologia , Neurônios/citologia , Retroviridae/genética , Telomerase/genética , Meios de Cultura , Humanos , Cariotipagem , Puromicina , Medula Espinal/citologia , Medula Espinal/embriologia , Telomerase/metabolismo , Telômero/fisiologia , Transfecção
20.
Proc Natl Acad Sci U S A ; 104(20): 8334-9, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17488820

RESUMO

Bub1 is a kinase believed to function primarily in the mitotic spindle checkpoint. Mutation or aberrant Bub1 expression is associated with chromosomal instability, aneuploidy, and human cancer. We now find that targeting Bub1 by RNAi or simian virus 40 (SV40) large T antigen in normal human diploid fibroblasts results in premature senescence. Interestingly, cells undergoing replicative senescence were also low in Bub1 expression, although ectopic Bub1 expression in presenescent cells was insufficient to extend lifespan. Premature senescence caused by lower Bub1 levels depends on p53. Senescence induction was blocked by dominant negative p53 expression or depletion of p21(CIP1), a p53 target. Importantly, cells with lower Bub1 levels and inactivated p53 became highly aneuploid. Taken together, our data highlight a role for p53 in monitoring Bub1 function, which may be part of a more general spindle checkpoint surveillance mechanism. Our data support the hypothesis that Bub1 compromise triggers p53-dependent senescence, which limits the production of aneuploid and potentially cancerous cells.


Assuntos
Proteínas Quinases/deficiência , Proteína Supressora de Tumor p53/metabolismo , Aneuploidia , Antígenos Virais de Tumores/metabolismo , Linhagem Celular , Proliferação de Células , Senescência Celular , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Ligação Proteica , Proteínas Serina-Treonina Quinases , Proteína do Retinoblastoma/metabolismo , Vírus 40 dos Símios/imunologia , Fuso Acromático/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...