Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Intervalo de año de publicación
1.
Nat Commun ; 15(1): 3873, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719882

RESUMEN

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.


Asunto(s)
Envejecimiento , MicroARNs , Neuroglía , Factores de Transcripción , Humanos , Neuroglía/metabolismo , Neuroglía/citología , Envejecimiento/genética , Envejecimiento/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , MicroARNs/genética , MicroARNs/metabolismo , Senescencia Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre/metabolismo , Células Madre/citología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Adulto , Redes Reguladoras de Genes , Proliferación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Perfilación de la Expresión Génica
2.
Carcinogenesis ; 40(7): 924-935, 2019 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-31155639

RESUMEN

The conditions that lead to antitumor or protumor functions of natural killer T (NKT) cells against mammalian tumors are only partially understood. Therefore, insights into the evolutionary conservation of NKT and their analogs-innate-like T (iT) cells-may reveal factors that contribute to tumor eradication. As such, we investigated the amphibian Xenopus laevis iT cells and interacting MHC class I-like (XNC or mhc1b.L) genes against ff-2 thymic lymphoid tumors. Upon ff-2 intraperitoneal transplantation into syngeneic tadpoles, two iT cell subsets iVα6 and iVα22, characterized by an invariant T-cell receptor α chain rearrangement (Vα6-Jα1.43 and Vα22-Jα1.32 respectively), were recruited to the peritoneum, concomitant with a decreased level of these transcripts in the spleen and thymus. To address the hypothesize that different iT cell subsets have distinct, possibly opposing, roles upon ff-2 tumor challenge, we determined whether ff-2 tumor growth could be manipulated by impairing Vα6 iT cells or by deleting their restricting element, the XNC gene, XNC10 (mhc1b10.1.L), on ff-2 tumors. Accordingly, the in vivo depletion of Vα6 iT cells using XNC10-tetramers enhanced tumor growth, indicating Vα6 iT cell-mediated antitumor activities. However, XNC10-deficient transgenic tadpoles that also lack Vα6 iT cells were resistant to ff-2 tumors, uncovering a potential new function of XNC10 besides Vα6 iT cell development. Furthermore, the CRISPR/Cas9-mediated knockout of XNC10 in ff-2 tumors broke the immune tolerance. Together, our findings demonstrate the relevance of XNC10/iT cell axis in controlling Xenopus tumor tolerance or rejection.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Células T Asesinas Naturales/inmunología , Subgrupos de Linfocitos T/inmunología , Neoplasias del Timo/inmunología , Escape del Tumor/inmunología , Proteínas de Xenopus/metabolismo , Animales , Animales Modificados Genéticamente , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Larva , Células T Asesinas Naturales/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Subgrupos de Linfocitos T/metabolismo , Neoplasias del Timo/patología , Proteínas de Xenopus/inmunología , Xenopus laevis
3.
Sci Signal ; 11(515)2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29382785

RESUMEN

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.


Asunto(s)
Astrocitos/metabolismo , Inhibidores Enzimáticos/farmacología , Colorantes Fluorescentes/farmacología , Túbulos Renales Proximales/metabolismo , Miocitos Cardíacos/metabolismo , Neuronas/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Compuestos de Anilina/farmacología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Células Cultivadas , Fura-2/farmacología , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/enzimología , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Neuronas/efectos de los fármacos , Neuronas/enzimología , Potasio/metabolismo , Xantenos/farmacología
4.
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
5.
J Neurosci ; 34(50): 16594-604, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25505312

RESUMEN

Experimental advances in the study of neuroglia signaling have been greatly accelerated by the generation of transgenic mouse models. In particular, an elegant manipulation that interferes with astrocyte vesicular release of gliotransmitters via overexpression of a dominant-negative domain of vesicular SNARE (dnSNARE) has led to documented astrocytic involvement in processes that were traditionally considered strictly neuronal, including the sleep-wake cycle, LTP, cognition, cortical slow waves, depression, and pain. A key premise leading to these conclusions was that expression of the dnSNARE was specific to astrocytes. Inconsistent with this premise, we report here widespread expression of the dnSNARE transgene in cortical neurons. We further demonstrate that the activity of cortical neurons is reversibly suppressed in dnSNARE mice. These findings highlight the need for independent validation of astrocytic functions identified in dnSNARE mice and thus question critical evidence that astrocytes contribute to neurotransmission through SNARE-dependent vesicular release of gliotransmitters.


Asunto(s)
Regulación de la Expresión Génica , Neuronas/metabolismo , Proteínas SNARE/biosíntesis , Animales , Animales Recién Nacidos , Células Cultivadas , Electroencefalografía/métodos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas SNARE/genética , Fases del Sueño/fisiología
6.
Cell Rep ; 3(6): 2127-41, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23727239

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Glioma/genética , Glioma/patología , Células Madre Neoplásicas/patología , Neuroglía/patología , Neuroglía/fisiología , Adulto , Neoplasias Encefálicas/metabolismo , Diferenciación Celular/fisiología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Humanos , Persona de Mediana Edad , Neuroglía/metabolismo , Activación Transcripcional
7.
Cell Stem Cell ; 12(6): 787-99, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23746982

RESUMEN

Huntington's disease (HD) is a neurodegenerative disease characterized in part by the loss of striatopallidal medium spiny projection neurons (MSNs). Expression of BDNF and noggin via intracerebroventricular (ICV) delivery in an adenoviral vector triggers the addition of new neurons to the neostriatum. In this study, we found that a single ICV injection of the adeno-associated viruses AAV4-BDNF and AAV4-noggin triggered the sustained recruitment of new MSNs in both wild-type and R6/2 mice, a model of HD. Mice treated with AAV4-BDNF/noggin or with BDNF and noggin proteins actively recruited subependymal progenitor cells to form new MSNs that matured and achieved circuit integration. Importantly, the AAV4-BDNF/noggin-treated R6/2 mice showed delayed deterioration of motor function and substantially increased survival. In addition, squirrel monkeys given ICV injections of adenoviral BDNF/noggin showed similar addition of striatal neurons. Induced neuronal addition may therefore represent a promising avenue for disease amelioration in HD.


Asunto(s)
Modelos Animales de Enfermedad , Progresión de la Enfermedad , Enfermedad de Huntington/patología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Animales , Ratones , Ratones Transgénicos
8.
Neurotherapeutics ; 8(4): 577-90, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21971961

RESUMEN

Huntington's disease (HD) is an inherited, relentlessly progressive neurodegenerative disease with an invariably fatal outcome. HD is inherited in an autosomal dominant fashion, and is characterized pathologically by the loss of cortical and striatal neurons, and clinically by involuntary choreiform movements accompanied by progressive cognitive impairment and emotional lability. The disorder is caused by an expanded cystosine adenine guanine (CAG) tri-nucleotide repeat encoding polyglutamine (polyQ) in the first exon of the Huntingtin gene. There is a correlation between the number of CAG repeats and disease onset, such that in patients with CAG repeat lengths of 36 to 60, disease symptoms typically manifest after 35 years of age, whereas CAG repeat lengths >60 yield the more severe juvenile form of the disease. Even though mutant huntingtin is expressed throughout the brain, it is characterized by the selective degeneration of medium spiny neurons of the caudate and putamen, which heralds more widespread neuronal degeneration with disease progression. The mechanisms of cell dysfunction and death in HD have been the subjects of a number of studies, which have led to therapeutic strategies largely based on the amelioration of mutant huntingtin-related metabolic impairment and cellular toxicity. Each of these approaches has aimed to delay or stop the preferential degeneration of medium spiny neurons early in the disease course. Yet, in later stages of the disease, after cell death has become prominent, cell replacement therapy (whether by direct cell transplantation or by the mobilization of endogenous progenitors) may comprise a stronger potential avenue for therapy. In this review, we will consider recent progress in the transplantation of fetal striatal cells to the HD brain, as well as emerging alternative sources for human striatal progenitor cells. We will then consider the potential application of gene therapy toward the induction of striatal neurogenesis and neuronal recruitment, with an eye toward its potential therapeutic use in HD.


Asunto(s)
Trasplante de Células/métodos , Enfermedad de Huntington/cirugía , Neuronas/fisiología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Encéfalo/citología , Encéfalo/embriología , Células Madre Embrionarias/fisiología , Células Madre Embrionarias/trasplante , Humanos , Enfermedad de Huntington/fisiopatología , Células Madre Pluripotentes/fisiología , Células Madre Pluripotentes/trasplante
9.
Mol Ther ; 18(1): 44-53, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19654569

RESUMEN

The development of Alzheimer's disease (AD) is closely connected with cholesterol metabolism. Cholesterol increases the production and deposition of amyloid-beta (Abeta) peptides that result in the formation of amyloid plaques, a hallmark of the pathology. In the brain, cholesterol is synthesized in situ but cannot be degraded nor cross the blood-brain barrier. The major exportable form of brain cholesterol is 24S-hydroxycholesterol, an oxysterol generated by the neuronal cholesterol 24-hydroxylase encoded by the CYP46A1 gene. We report that the injection of adeno-associated vector (AAV) encoding CYP46A1 in the cortex and hippocampus of APP23 mice before the onset of amyloid deposits markedly reduces Abeta peptides, amyloid deposits and trimeric oligomers at 12 months of age. The Morris water maze (MWM) procedure also demonstrated improvement of spatial memory at 6 months, before the onset of amyloid deposits. AAV5-wtCYP46A1 vector injection in the cortex and hippocampus of amyloid precursor protein/presenilin 1 (APP/PS) mice after the onset of amyloid deposits also reduced markedly the number of amyloid plaques in the hippocampus, and to a less extent in the cortex, 3 months after the injection. Our data demonstrate that neuronal overexpression of CYP46A1 before or after the onset of amyloid plaques significantly reduces Abeta pathology in mouse models of AD.


Asunto(s)
Enfermedad de Alzheimer/terapia , Amiloide/metabolismo , Dependovirus/genética , Terapia Genética/métodos , Esteroide Hidroxilasas/fisiología , Enfermedad de Alzheimer/metabolismo , Animales , Western Blotting , Línea Celular , Colesterol 24-Hidroxilasa , Ensayo de Inmunoadsorción Enzimática , Humanos , Hidroxicolesteroles/metabolismo , Inmunohistoquímica , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esteroide Hidroxilasas/genética
10.
J Clin Invest ; 117(10): 2889-902, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17885687

RESUMEN

Ependymal overexpression of brain-derived neurotrophic factor (BDNF) stimulates neuronal addition to the adult striatum, from subependymal progenitor cells. Noggin, by suppressing subependymal gliogenesis and increasing progenitor availability, potentiates this process. We asked whether BDNF/Noggin overexpression might be used to recruit new striatal neurons in R6/2 huntingtin transgenic mice. R6/2 mice injected with adenoviral BDNF and adenoviral Noggin (AdBDNF/AdNoggin) recruited BrdU(+)betaIII-tubulin(+) neurons, which developed as DARPP-32(+) and GABAergic medium spiny neurons that expressed either enkephalin or substance P and extended fibers to the globus pallidus. Only AdBDNF/AdNoggin-treated R6/2 mice harbored migrating doublecortin-defined neuroblasts in their striata, and the new neurons expressed p27 as a marker of mitotic quiescence after parenchymal integration. AdBDNF/AdNoggin-treated R6/2 mice sustained their rotarod performance and open-field activity and survived longer than did AdNull-treated and untreated controls. Neither motor performance nor survival improved in R6/2 mice treated only with AdBDNF, and intraventricular infusion of the mitotic inhibitor Ara-C completely blocked the performance and survival effects of AdBDNF/AdNoggin, suggesting that the benefits of AdBDNF/AdNoggin derived from neuronal addition. Thus, BDNF and Noggin induced striatal neuronal regeneration, delayed motor impairment, and extended survival in R6/2 mice, suggesting a new therapeutic strategy in Huntington disease.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Proteínas Portadoras/genética , Enfermedad de Huntington/terapia , Neostriado/fisiología , Neuronas/fisiología , Regeneración , Adenoviridae/genética , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalinas/análisis , Encefalinas/metabolismo , Globo Pálido/citología , Globo Pálido/fisiología , Enfermedad de Huntington/fisiopatología , Ratones , Ratones Transgénicos , Mitosis , Neostriado/citología , Neuronas/química , Neuronas/metabolismo , Sustancia P/análisis , Sustancia P/metabolismo , Tubulina (Proteína)/metabolismo
11.
Hum Mol Genet ; 15(1): 53-64, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16311251

RESUMEN

Metachromatic leukodystrophy (MLD) is a neurodegenerative lysosomal disease caused by a defect of the enzyme arylsulfatase A (ARSA) that disrupts the degradation of sulfatides (Sulf) in neurons and glial cells. Therapy for MLD requires active production of ARSA in the brain to prevent demyelination and neuronal damage, and efficient delivery of ARSA to act faster than disease progression, particularly in the rapidly progressive late infantile form. We used an adeno-associated virus serotype 5 (AAV5) vector to express the human ARSA gene in the brain of MLD mouse model. We achieved rapid, extensive and long-lasting expression of the recombinant ARSA in the brain, cerebellum and brainstem from at least 3 to 15 months post-injection. Analysis of the vector genome and ARSA distribution gave evidence for in vivo cross-correction of many untransduced neurons and astrocytes. ARSA delivery rapidly reversed Sulf storage and prevented neuropathological abnormalities and neuromotor impairment. We believe that AAV5-mediated brain delivery of ARSA is a potentially efficacious therapeutic strategy for MLD patients, especially for those with rapidly progressive form of the disease.


Asunto(s)
Encéfalo/metabolismo , Cerebrósido Sulfatasa/metabolismo , Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Leucodistrofia Metacromática/terapia , Adenoviridae , Análisis de Varianza , Animales , Encéfalo/patología , Cerebrósido Sulfatasa/genética , Electrofisiología , Vectores Genéticos/genética , Humanos , Inmunohistoquímica , Leucodistrofia Metacromática/enzimología , Ratones , Ratones Mutantes , Prueba de Desempeño de Rotación con Aceleración Constante
12.
Exp Cell Res ; 312(3): 233-44, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16300754

RESUMEN

Notch signaling participates in the development of multicellular organisms by maintaining self-renewal potential or inducing differentiation of numerous tissues. In this study, we characterized Notch4, the evolutionary most distant and least studied Notch family member. We identified a Notch4 inter-strain polymorphism with a previously undescribed mRNA variant. This longer Notch4 mRNA, which represented up to one-third of total Notch4 mRNA, resulted from intron 10 retention. Analysis of Notch4 intron 10 revealed that an 8-bp deletion, reducing its length from 68 to 60 bp, strictly correlated with its retention. Further experiments demonstrated that intron length was the only cause of the mis-splicing. Moreover, this mRNA variant resulted in a truncated protein containing half the extracellular domain of Notch4, including the ligand-binding domain.


Asunto(s)
Intrones/genética , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , Receptores Notch/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Complejo Mayor de Histocompatibilidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Polimorfismo Genético , Proteínas Proto-Oncogénicas/biosíntesis , Empalme del ARN , Receptor Notch4 , Receptores Notch/biosíntesis , Eliminación de Secuencia
13.
J Gene Med ; 6(5): 555-64, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15133766

RESUMEN

BACKGROUND: The development of stable producer cell lines for recombinant adeno-associated virus (rAAV) assembly is a strategy followed by many groups to develop scalable production methods suitable for good manufacturing practice (GMP) requirements. The major drawback of this method lies in the requirement for replicating adenovirus (Ad) for rAAV assembly. In the present study, we analyzed the ability of several replication-defective herpes simplex type 1 (HSV-1) helper viruses to induce rAAV2 particle production from stable producer cell lines. METHODS: Several stable rAAV producer cell clones were infected with wild-type and replication-defective HSV strains and analyzed for rep-cap gene amplification, viral protein synthesis and rAAV titers achieved. In vivo analysis following rAAV injection in the murine brain was also conducted to evaluate the toxicity and biopotency of the rAAV stocks. RESULTS: We demonstrated that an HSV strain mutated in the UL30 polymerase gene could efficiently be used in this context, resulting in rAAV titers similar to those measured with wild-type HSV or Ad. Importantly, with respect to clinical developments, the use of this mutant resulted in rAAV stocks which were consistently devoid of contaminating HSV particles and fully active in vivo in the murine central nervous system with no detectable toxicity. CONCLUSIONS: This study, together with our previous report describing a rAAV chromatography-based purification process, contributes to the definition of an entirely scalable process for the generation of rAAV particles.


Asunto(s)
Dependovirus/fisiología , Herpesvirus Humano 1/fisiología , Replicación Viral , Animales , Chlorocebus aethiops , Dependovirus/genética , Femenino , Vectores Genéticos , Células HeLa , Herpesvirus Humano 1/genética , Humanos , Ratones , Mutación , Recombinación Genética , Células Vero , Ensamble de Virus
14.
J Neurosci ; 24(9): 2133-42, 2004 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-14999064

RESUMEN

Neurogenesis from endogenous progenitor cells in the adult forebrain ventricular wall may be induced by the local viral overexpression of cognate neuronal differentiation agents, in particular BDNF. Here, we show that the overexpression of noggin, by acting to inhibit glial differentiation by subependymal progenitor cells, can potentiate adenoviral BDNF-mediated recruitment of new neurons to the adult rat neostriatum. The new neurons survive at least 2 months after their genesis in the subependymal zone and are recruited primarily as GABAergic DARPP-32+ medium spiny neurons in the caudate-putamen. The new medium spiny neurons successfully project to the globus pallidus, their usual developmental target, extending processes over several millimeters of the normal adult striatum. Thus, concurrent suppression of subependymal glial differentiation and promotion of neuronal differentiation can mobilize endogenous subependymal progenitor cells to achieve substantial neuronal addition to otherwise non-neurogenic regions of the adult brain.


Asunto(s)
Adenoviridae/genética , Proteínas Morfogenéticas Óseas/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Neuronas/metabolismo , Células Madre/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/farmacología , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/farmacología , Bromodesoxiuridina , Proteínas Portadoras , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Colorantes Fluorescentes , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Globo Pálido/citología , Humanos , Ventrículos Laterales/citología , Ventrículos Laterales/efectos de los fármacos , Neostriado/citología , Neostriado/efectos de los fármacos , Neuroglía/citología , Neuroglía/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Células Madre/citología , Células Madre/efectos de los fármacos , Estilbamidinas
15.
J Neurosci Res ; 69(6): 966-75, 2002 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12205690

RESUMEN

A distinct population of white matter progenitor cells (WMPCs), competent but not committed to generate oligodendrocytes, remains ubiquitous in the adult human subcortical white matter. These cells are present in both sexes and into senescence and may constitute as much as 4% of the cells of adult human capsular white matter. Transduction of adult human white matter dissociates with plasmids bearing early oligodendrocytic promoters driving fluorescent reporters permits the separation of these cells at high yield and purity, as does separation based on their expression of A2B5 immunoreactivity. Isolates of these cells survive xenograft to lysolecithin-demyelinated brain and migrate rapidly to infiltrate these lesions, without extending into normal white matter. Within several weeks, implanted progenitors mature as oligodendrocytes, and develop myelin-associated antigens. Lentiviral tagging with green fluorescent protein confirmed that A2B5-sorted progenitors develop myelin basic protein expression within regions of demyelination and that they fail to migrate when implanted into normal brain. Adult human white matter progenitor cells can thus disperse widely through regions of experimental demyelination and are able to differentiate as myelinating oligodendrocytes. This being the case, they may constitute appropriate vectors for cell-based remyelination strategies.


Asunto(s)
Trasplante de Tejido Encefálico , Enfermedades Desmielinizantes/cirugía , Oligodendroglía/citología , Trasplante de Células Madre , Células Madre/citología , Adulto , Factores de Edad , Animales , Diferenciación Celular , Movimiento Celular , Supervivencia Celular , Femenino , Expresión Génica , Vectores Genéticos , Supervivencia de Injerto , Proteínas Fluorescentes Verdes , Humanos , Separación Inmunomagnética , Indicadores y Reactivos/metabolismo , Lentivirus/genética , Proteínas Luminiscentes/genética , Lisofosfatidilcolinas , Masculino , Persona de Mediana Edad , Proteína Básica de Mielina/análisis , Vaina de Mielina , Neuronas/citología , Oligodendroglía/química , Ratas , Trasplante Heterólogo
16.
Biochim Biophys Acta ; 1583(1): 45-52, 2002 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-12069848

RESUMEN

Stearoyl-CoA desaturase (SCD) is a short-lived integral membrane protein of the endoplasmic reticulum (ER) that catalyzes the insertion of a double bond in the delta 9 position of saturated fatty acids. Its expression has been difficult in heterologous systems. In this study, recombinant adenovirus vector was used to express both wild-type (wt) and engineered forms of rat SCD in human transformed kidney cells. In the engineered form of SCD, lysyl residues at positions 33, 35, and 36 were mutated to alanine (SCD K/A). The recombinant adenovirus also contains a cDNA encoding the green fluorescent protein (GFP). The stable reporter GFP was used to analyze the efficiency of transfection and the stability of expressed SCDs. The wt SCD was unstable upon expression, whereas expression of SCD K/A resulted in the stabilization of the protein. The proteasome inhibitor MG132 did not affect the rapid degradation of expressed wt SCD, implying that proteasome is not involved in this degradation. Functional analysis of microsomes from infected cells expressing SCD K/A resulted in the formation of holoenzyme with desaturase activity. Here we report engineering a stabilized form of a rapidly degraded membrane protein for production of an active mutant form of SCD. The adenovirus transformed cells may provide a model for the study of the effects of positive SCD expression.


Asunto(s)
Ácido Graso Desaturasas/metabolismo , Lisina/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Cisteína Endopeptidasas/metabolismo , Cartilla de ADN , Estabilidad de Enzimas , Ácido Graso Desaturasas/genética , Humanos , Complejos Multienzimáticos/metabolismo , Mutagénesis Sitio-Dirigida , Complejo de la Endopetidasa Proteasomal , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estearoil-CoA Desaturasa , Ubiquitina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA