Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Brain Res ; 1754: 147254, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33422542

RESUMEN

Schizophrenia is a neurodevelopmental psychiatric disorder, encompassing genetic and environmental risk factors. For several decades, investigators have been implementing the use of lesions of the neonatal rodent hippocampus to model schizophrenia, resulting in a broad spectrum of adult schizophrenia-related behavioral changes. Despite the extensive use of these proposed animal models of schizophrenia, the mechanisms by which these lesions result in schizophrenia-like behavioral alterations remain unclear. Here we provide in vivo evidence that transient pharmacological inactivation of the hippocampus via tetrodotoxin microinjections or a genetic reduction in brain derived neurotrophic factor (BDNF) protein levels (BDNF+/- rats) lead to global DNA hypomethylation, disrupted maturation of the neuronal nucleus and aberrant acoustic startle response in the adult rat. The similarity between the effects of the two treatments strongly indicate that BDNF signaling is involved in effects obtained after the TTX microinjections. These findings may shed light on the cellular mechanisms underlying the phenotypical features of neonatal transient inhibition of the hippocampus as a preclinical model of schizophrenia and suggest that BDNF signaling represents a target pathway for development of novel treatment therapies.


Asunto(s)
Conducta Animal/fisiología , Factor Neurotrófico Derivado del Encéfalo/deficiencia , Metilación de ADN/fisiología , ADN/metabolismo , Hipocampo/metabolismo , Animales , Animales Recién Nacidos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neuronas/metabolismo , Ratas , Reflejo de Sobresalto/genética , Reflejo de Sobresalto/fisiología , Esquizofrenia/genética , Esquizofrenia/metabolismo
2.
Nat Commun ; 11(1): 462, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31974374

RESUMEN

Human induced pluripotent stem cells (hiPSCs) are a powerful model of neural differentiation and maturation. We present a hiPSC transcriptomics resource on corticogenesis from 5 iPSC donor and 13 subclonal lines across 9 time points over 5 broad conditions: self-renewal, early neuronal differentiation, neural precursor cells (NPCs), assembled rosettes, and differentiated neuronal cells. We identify widespread changes in the expression of both individual features and global patterns of transcription. We next demonstrate that co-culturing human NPCs with rodent astrocytes results in mutually synergistic maturation, and that cell type-specific expression data can be extracted using only sequencing read alignments without cell sorting. We lastly adapt a previously generated RNA deconvolution approach to single-cell expression data to estimate the relative neuronal maturity of iPSC-derived neuronal cultures and human brain tissue. Using many public datasets, we demonstrate neuronal cultures are maturationally heterogeneous but contain subsets of neurons more mature than previously observed.


Asunto(s)
Diferenciación Celular/genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología , Células-Madre Neurales/fisiología , Transcriptoma , Algoritmos , Animales , Astrocitos/citología , Células Cultivadas , Corteza Cerebral/citología , Técnicas de Cocultivo , Bases de Datos Genéticas , Regulación de la Expresión Génica , Humanos , Modelos Neurológicos , Células-Madre Neurales/citología , Neuronas/citología , Neuronas/fisiología , Ratas
3.
Artículo en Inglés | MEDLINE | ID: mdl-30568848

RESUMEN

The cerebellum, a derivative of the hindbrain, plays a crucial role in balance and posture as well as in higher cognitive and locomotive processes. Cerebellar development is initiated during the segmental phase of hindbrain formation. Here, we describe the phenotype, of a single surviving adult conditional mouse mutant mouse, in which Sox2 function is ablated in embryonic radial glial cells by means of hGFAP-CRE. The single Sox2RGINV/mosaic adult mutant mouse displays motor disability, microsomia, reduced Central Nervous System (CNS) size and cerebellar defects associated with human genetically related congenital abnormalities.

4.
Bioorg Med Chem Lett ; 28(19): 3231-3235, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30170942

RESUMEN

Due to increased interest in As(III) S-adenosylmethionine methyltransferase (AS3MT), a search for chemical probes that can help elucidate function was initiated. A homology model was built based on related enzymes, and virtual screening produced 426 potential hits. Evaluation of these compounds in a functional enzymatic assay revealed several modest inhibitors including an O-substituted 2-amino-3-cyano indole scaffold. Two iterations of near neighbor searches revealed compound 5 as a potent inhibitor of AS3MT with good selectivity over representative methyltransferases DOT1L and NSD2 as well as a representative set of diverse receptors. Compound 5 should prove to be a useful tool to investigate the role of AS3MT and a potential starting point for further optimization.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Metiltransferasas/antagonistas & inhibidores , Humanos
5.
Trends Mol Med ; 24(9): 805-820, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30006147

RESUMEN

Use of human pluripotent stem cells (hPSCs) and their differentiated derivatives have led to recent proof-of-principle drug discoveries, defining a pathway to the implementation of hPSC-based drug discovery (hPDD). Current hPDD strategies, however, have inevitable conceptual biases and technological limitations, including the dimensionality of cell-culture methods, cell maturity and functionality, experimental variability, and data reproducibility. In this review, we dissect representative hPDD systems via analysis of hPSC-based 2D-monolayers, 3D culture, and organoids. We discuss mechanisms of drug discovery and drug repurposing, and roles of membrane drug transporters in tissue maturation and hPDD using the example of drugs that target various mutations of CFTR, the cystic fibrosis transmembrane conductance regulator gene, in patients with cystic fibrosis.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Desarrollo de Medicamentos/métodos , Descubrimiento de Drogas/métodos , Células Madre Pluripotentes/efectos de los fármacos , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Técnicas de Cultivo de Célula/instrumentación , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Desarrollo de Medicamentos/instrumentación , Descubrimiento de Drogas/instrumentación , Humanos , Terapia Molecular Dirigida/métodos , Organoides/citología , Organoides/efectos de los fármacos , Organoides/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
6.
Stem Cells ; 36(1): 11-21, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28948674

RESUMEN

Lineage commitment and differentiation of skeletal stem cells/bone marrow stromal cells (SSCs/BMSCs, often called bone marrow-derived "mesenchymal stem/stromal" cells) offer an important opportunity to study skeletal and hematopoietic diseases, and for tissue engineering and regenerative medicine. Currently, many studies in this field have relied on cell lineage tracing methods in mouse models, which have provided a significant advancement in our knowledge of skeletal and hematopoietic stem-cell niches in bone marrow (BM). However, there is a lack of agreement in numerous fundamental areas, including origins of various BM stem-cell niches, cell identities, and their physiological roles in the BM. In order to resolve these issues, we propose a new hypothesis of "paralogous" stem-cell niches (PSNs); that is, progressively altered parallel niches within an individual species throughout the life span of the organism. A putative PSN code seems to be plausible based on analysis of transcriptional signatures in two representative genes that encode Nes-GFP and leptin receptors, which are frequently used to monitor SSC lineage development in BM. Furthermore, we suggest a dynamic paralogous BM niche (PBMN) model that elucidates the coupling and uncoupling mechanisms between BM stem-cell niches and their zones of active regeneration during different developmental stages. Elucidation of these PBMNs would enable us to resolve the existing controversies, thus paving the way to achieving precision regenerative medicine and pharmaceutical applications based on these BM cell resources. Stem Cells 2018;36:11-21.


Asunto(s)
Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Nicho de Células Madre/genética , Células Madre/metabolismo , Diferenciación Celular , Linaje de la Célula , Humanos
7.
Front Physiol ; 7: 127, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27148066

RESUMEN

Temporal and spatial control of gene expression can be achieved using an inducible system as a fundamental tool for regulated transcription in basic, applied and eventually in clinical research. We describe a novel "hit and run" inducible direct reprogramming approach. In a single step, 2 days post-transfection, transiently transfected Sox2(FLAG) under the Leu3p-αIPM inducible control (iSox2) triggers the activation of endogenous Sox2, redirecting primary astrocytes into abundant distinct nestin-positive radial glia cells. This technique introduces a unique novel tool for safe, rapid and efficient reprogramming amendable to regenerative medicine.

8.
Stem Cells Transl Med ; 3(7): 867-78, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24855277

RESUMEN

The ability to differentiate induced pluripotent stem cells (iPSCs) into committed skeletal progenitors could allow for an unlimited autologous supply of such cells for therapeutic uses; therefore, we attempted to create novel bone-forming cells from human iPSCs using lines from two distinct tissue sources and methods of differentiation that we previously devised for osteogenic differentiation of human embryonic stem cells, and as suggested by other publications. The resulting cells were assayed using in vitro methods, and the results were compared with those obtained from in vivo transplantation assays. Our results show that true bone was formed in vivo by derivatives of several iPSC lines, but that the successful cell lines and differentiation methodologies were not predicted by the results of the in vitro assays. In addition, bone was formed equally well from iPSCs originating from skin or bone marrow stromal cells (also known as bone marrow-derived mesenchymal stem cells), suggesting that the iPSCs did not retain a "memory" of their previous life. Furthermore, one of the iPSC-derived cell lines formed verifiable cartilage in vivo, which likewise was not predicted by in vitro assays.


Asunto(s)
Bioensayo/métodos , Diferenciación Celular , Condrocitos/metabolismo , Condrogénesis , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Anciano , Anciano de 80 o más Años , Animales , Línea Celular , Reprogramación Celular , Condrocitos/trasplante , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Masculino , Trasplante de Células Madre Mesenquimatosas , Ratones , Osteoblastos/trasplante , Fenotipo , Transfección
9.
Stem Cell Res ; 12(3): 610-21, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24603366

RESUMEN

Human pluripotent stem cells (hPSCs) have two potentially attractive applications: cell replacement-based therapies and drug discovery. Both require the efficient generation of large quantities of clinical-grade stem cells that are free from harmful genomic alterations. The currently employed colony-type culture methods often result in low cell yields, unavoidably heterogeneous cell populations, and substantial chromosomal abnormalities. Here, we shed light on the structural relationship between hPSC colonies/embryoid bodies and early-stage embryos in order to optimize current culture methods based on the insights from developmental biology. We further highlight core signaling pathways that underlie multiple epithelial-to-mesenchymal transitions (EMTs), cellular heterogeneity, and chromosomal instability in hPSCs. We also analyze emerging methods such as non-colony type monolayer (NCM) and suspension culture, which provide alternative growth models for hPSC expansion and differentiation. Furthermore, based on the influence of cell-cell interactions and signaling pathways, we propose concepts, strategies, and solutions for production of clinical-grade hPSCs, stem cell precursors, and miniorganoids, which are pivotal steps needed for future clinical applications.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Mamíferos/embriología , Células Madre Pluripotentes/citología , Transducción de Señal , Animales , Humanos , Mamíferos/genética , Mamíferos/metabolismo , Células Madre Pluripotentes/metabolismo
10.
Cell Stem Cell ; 14(1): 13-26, 2014 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-24388173

RESUMEN

Human pluripotent stem cells (hPSCs) provide powerful resources for application in regenerative medicine and pharmaceutical development. In the past decade, various methods have been developed for large-scale hPSC culture that rely on combined use of multiple growth components, including media containing various growth factors, extracellular matrices, 3D environmental cues, and modes of multicellular association. In this Protocol Review, we dissect these growth components by comparing cell culture methods and identifying the benefits and pitfalls associated with each one. We further provide criteria, considerations, and suggestions to achieve optimal cell growth for hPSC expansion, differentiation, and use in future therapeutic applications.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/trasplante , Diferenciación Celular , Humanos
11.
Stem Cells ; 32(3): 770-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24155149

RESUMEN

The ex vivo expansion of stem cells is making major contribution to biomedical research. The multipotent nature of neural precursors acutely isolated from the developing central nervous system has been established in a series of studies. Understanding the mechanisms regulating cell expansion in tissue culture would support their expanded use either in cell therapies or to define disease mechanisms. Basic fibroblast growth factor (FGF2) and insulin, ligands for tyrosine kinase receptors, are sufficient to sustain neural stem cells (NSCs) in culture. Interestingly, real-time imaging shows that these cells become multipotent every time they are passaged. Here, we analyze the role of FGF2 and insulin in the brief period when multipotent cells are present. FGF2 signaling results in the phosphorylation of Erk1/2, and activation of c-Fos and c-Jun that lead to elevated cyclin D mRNA levels. Insulin signals through the PI3k/Akt pathway to regulate cyclins at the post-transcriptional level. This precise Boolean regulation extends our understanding of the proliferation of multipotent NSCs and provides a basis for further analysis of proliferation control in the cell states defined by real-time mapping of the cell lineages that form the central nervous system.


Asunto(s)
Ciclina D/genética , Factor 2 de Crecimiento de Fibroblastos/farmacología , Insulina/farmacología , Células Madre Multipotentes/citología , Células-Madre Neurales/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Ciclina D/metabolismo , ADN/biosíntesis , Femenino , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/enzimología , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transcripción Genética/efectos de los fármacos
12.
Stem Cell Res ; 10(1): 57-66, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23117585

RESUMEN

Much of the excitement generated by induced pluripotent stem cell technology is concerned with the possibility of disease modeling as well as the potential for personalized cell therapy. However, to pursue this it is important to understand the 'normal' pluripotent state including its inherent variability. We have performed various molecular profiling assays for 21 hESC lines and 8 hiPSC lines to generate a comprehensive snapshot of the undifferentiated state of pluripotent stem cells. Analysis of the gene expression data revealed no iPSC-specific gene expression pattern in accordance with previous reports. We further compared cells, differentiated as embryoid bodies in 2 media proposed to initiate differentiation towards separate cell fates, as well as 20 adult tissues. From this analysis we have generated a gene list which defines pluripotency and establishes a baseline for the pluripotent state. Finally, we provide lists of genes enriched under both differentiation conditions which show the proposed bias toward independent cell fates.


Asunto(s)
Bases de Datos Factuales , Células Madre Pluripotentes/metabolismo , Animales , Línea Celular , Perfilación de la Expresión Génica , Humanos , Ratones , National Institutes of Health (U.S.) , Células Madre Pluripotentes/citología , Análisis de Componente Principal , Estados Unidos
13.
Stem Cells ; 30(10): 2175-87, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22887864

RESUMEN

The expression and function of several multidrug transporters (including ABCB1 and ABCG2) have been studied in human cancer cells and in mouse and human adult stem cells. However, the expression of ABCG2 in human embryonic stem cells (hESCs) remains unclear. Limited and contradictory results in the literature from two research groups have raised questions regarding its expression and function. In this study, we used quantitative real-time PCR, Northern blots, whole genome RNA sequencing, Western blots, and immunofluorescence microscopy to study ABCG2 expression in hESCs. We found that full-length ABCG2 mRNA transcripts are expressed in undifferentiated hESC lines. However, ABCG2 protein was undetectable even under embryoid body differentiation or cytotoxic drug induction. Moreover, surface ABCG2 protein was coexpressed with the differentiation marker stage-specific embryonic antigen-1 of hESCs, following constant BMP-4 signaling at days 4 and 6. This expression was tightly correlated with the downregulation of two microRNAs (miRNAs) (i.e., hsa-miR-519c and hsa-miR-520h). Transfection of miRNA mimics and inhibitors of these two miRNAs confirmed their direct involvement in the regulation ABCG2 translation. Our findings clarify the controversy regarding the expression of the ABCG2 gene and also provide new insights into translational control of the expression of membrane transporter mRNAs by miRNAs in hESCs.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , MicroARNs/genética , Proteínas de Neoplasias/genética , ARN Mensajero/biosíntesis , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Nutrientes , Fibroblastos , Humanos , Antígeno Lewis X/genética , Antígeno Lewis X/metabolismo , Ratones , MicroARNs/metabolismo , Proteínas de Neoplasias/metabolismo , Biosíntesis de Proteínas , Transfección
14.
Stem Cell Res ; 9(3): 237-48, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22910561

RESUMEN

Regenerative medicine, relying on human embryonic stem cell (hESC) technology, opens promising new avenues for therapy of many severe diseases. However, this approach is restricted by limited production of the desired cells due to the refractory properties of hESC growth in vitro. It is further hindered by insufficient control of cellular stress, growth rates, and heterogeneous cellular states under current culture conditions. In this study, we report a novel cell culture method based on a non-colony type monolayer (NCM) growth. Human ESCs under NCM remain pluripotent as determined by teratoma assays and sustain the potential to differentiate into three germ layers. This NCM culture has been shown to homogenize cellular states, precisely control growth rates, significantly increase cell production, and enhance hESC recovery from cryopreservation without compromising chromosomal integrity. This culture system is simple, robust, scalable, and suitable for high-throughput screening and drug discovery.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Proliferación Celular , Células Madre Embrionarias/citología , Diferenciación Celular , Línea Celular , Expresión Génica , Humanos
15.
Gastroenterology ; 142(3): 602-11, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22138358

RESUMEN

BACKGROUND & AIMS: Many studies of embryonic stem cells have investigated direct cell replacement of damaged tissues, but little is known about how donor cell-derived signals affect host tissue regeneration. We investigated the direct and indirect roles of human embryonic stem cell-derived cells in liver repair in mice. METHODS: To promote the initial differentiation of human embryonic stem cells into mesendoderm, we activated the ß-catenin signaling pathway with lithium; cells were then further differentiated into hepatocyte-like cells. The differentiated cells were purified by indocyanine green staining and laser microdissection and characterized by immunostaining, polymerase chain reaction, biochemical function, electron microscopy, and transplantation analyses. To investigate indirect effects of these cells, secreted proteins (secretomes) were analyzed by a label-free quantitative mass spectrometry. Carbon tetrachloride was used to induce acute liver injury in mice; cells or secreted proteins were administered by intrasplenic or intraperitoneal injection, respectively. RESULTS: The differentiated hepatocyte-like cells had multiple features of normal hepatocytes, engrafted efficiently into mice, and continued to have hepatic features; they promoted proliferation of host hepatocytes and revascularization of injured host liver tissues. Proteomic analysis identified proteins secreted from these cells that might promote host tissue repair. Injection of the secreted proteins into injured livers of mice promoted significant amounts of tissue regeneration without cell grafts. CONCLUSIONS: Hepatocyte-like cells derived from human embryonic stem cells contribute to recovery of injured liver tissues in mice, not only by cell replacement but also by delivering trophic factors that support endogenous liver regeneration.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Enfermedad Hepática Inducida por Sustancias y Drogas/cirugía , Células Madre Embrionarias/trasplante , Hepatocitos/trasplante , Células Madre Pluripotentes Inducidas/trasplante , Regeneración Hepática , Hígado/patología , Animales , Biomarcadores/metabolismo , Tetracloruro de Carbono , Diferenciación Celular/efectos de los fármacos , Separación Celular/métodos , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Captura por Microdisección con Láser , Cloruro de Litio/farmacología , Hígado/irrigación sanguínea , Hígado/metabolismo , Espectrometría de Masas , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Electrónica , Neovascularización Fisiológica , Reacción en Cadena de la Polimerasa , Proteómica/métodos , Factores de Tiempo , Cicatrización de Heridas
16.
Cell Stem Cell ; 8(3): 318-25, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21362571

RESUMEN

Pluripotent stem cells provide a platform to interrogate control elements that function to generate all cell types of the body. Despite their utility for modeling development and disease, the relationship of mouse and human pluripotent stem cell states to one another remains largely undefined. We have shown that mouse embryonic stem (ES) cells and epiblast stem cells (EpiSCs) are distinct, pluripotent states isolated from pre- and post-implantation embryos respectively. Human ES cells are different than mouse ES cells and share defining features with EpiSCs, yet are derived from pre-implantation human embryos. Here we show that EpiSCs can be routinely derived from pre-implantation mouse embryos. The preimplantation-derived EpiSCs exhibit molecular features and functional properties consistent with bona fide EpiSCs. These results provide a simple method for isolating EpiSCs and offer direct insight into the intrinsic and extrinsic mechanisms that regulate the acquisition of distinct pluripotent states.


Asunto(s)
Blastocisto/citología , Separación Celular/métodos , Estratos Germinativos/citología , Células Madre/citología , Animales , Secuencia de Bases , Blastocisto/metabolismo , Diferenciación Celular/genética , Islas de CpG/genética , Metilación de ADN/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Células Madre/metabolismo
17.
PLoS One ; 5(5): e10841, 2010 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-20520777

RESUMEN

BACKGROUND: New mechanisms that regulate neural stem cell (NSC) expansion will contribute to improved assay systems and the emerging regenerative approach that targets endogenous stem cells. Expanding knowledge on the control of stem cell self renewal will also lead to new approaches for targeting the stem cell population of cancers. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that Cholera toxin regulates two recently characterized NSC markers, the Tie2 receptor and the transcription factor Hes3, and promotes the expansion of NSCs in culture. Cholera toxin increases immunoreactivity for the Tie2 receptor and rapidly induces the nuclear localization of Hes3. This is followed by powerful cultured NSC expansion and induction of proliferation both in the presence and absence of mitogen. CONCLUSIONS/SIGNIFICANCE: Our data suggest a new cell biological mechanism that regulates the self renewal and differentiation properties of stem cells, providing a new logic to manipulate NSCs in the context of regenerative disease and cancer.


Asunto(s)
Encéfalo/citología , Toxina del Cólera/farmacología , Feto/citología , Neuronas/citología , Transducción de Señal/efectos de los fármacos , Células Madre/citología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Receptor TIE-2/metabolismo , Proteínas Represoras , Células Madre/efectos de los fármacos , Células Madre/metabolismo
18.
Dev Growth Differ ; 52(3): 293-301, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20298258

RESUMEN

Gastrulation is the defining feature of metazoan development where it serves to apportion seemingly equivalent, pluripotent cells to specific fates. The three embryonic germ layers generated during gastrulation from the pluripotent epiblast including ectoderm, mesoderm, and definitive endoderm, contain the progenitors required to build all of the tissues of the developing organism. As a result, there is great interest in understanding the events that coordinate gastrulation. Because developing embryos in placental mammals are relatively inaccessible, stem cells are widely used for experimental and biochemical interrogation of these processes. Epiblast stem cells (EpiSCs) are grown from the post-implantation epiblast, which is the most proximal pluripotent tissue to the early somatic and germ cell precursors. Because EpiSCs can be propagated indefinitely in vitro as a stable state that recapitulates the properties of the post-implantation epiblast, they are uniquely positioned to provide novel insight into the developmental window where somatic and germ cell lineages are first established. Here we discuss the nature of EpiSCs and their significance in understanding gastrulation and cell specification in relationship to other pluripotent cell culture models.


Asunto(s)
Células Madre Embrionarias/citología , Estratos Germinativos/citología , Animales , Diferenciación Celular , Separación Celular , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Femenino , Gastrulación/genética , Gastrulación/fisiología , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/metabolismo , Humanos , Ratones , Modelos Biológicos , Proteína Nodal/genética , Proteína Nodal/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Embarazo , Primates , Transducción de Señal
19.
PLoS One ; 5(2): e9414, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20195471

RESUMEN

BACKGROUND: The ability to grow a uniform cell type from the adult central nervous system (CNS) is valuable for developing cell therapies and new strategies for drug discovery. The adult mammalian brain is a source of neural stem cells (NSC) found in both neurogenic and non-neurogenic zones but difficulties in culturing these hinders their use as research tools. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that NSCs can be efficiently grown in adherent cell cultures when angiogenic signals are included in the medium. These signals include both anti-angiogenic factors (the soluble form of the Notch receptor ligand, Dll4) and pro-angiogenic factors (the Tie-2 receptor ligand, Angiopoietin 2). These treatments support the self renewal state of cultured NSCs and expression of the transcription factor Hes3, which also identifies the cancer stem cell population in human tumors. In an organotypic slice model, angiogenic factors maintain vascular structure and increase the density of dopamine neuron processes. CONCLUSIONS/SIGNIFICANCE: We demonstrate new properties of adult NSCs and a method to generate efficient adult NSC cultures from various central nervous system areas. These findings will help establish cellular models relevant to cancer and regeneration.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Proliferación Celular/efectos de los fármacos , Neuronas/citología , Células Madre/citología , Adulto , Células Madre Adultas/citología , Angiopoyetina 2/farmacología , Animales , Encéfalo/citología , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Haplorrinos , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/farmacología , Ratones , Ratas , Proteínas Represoras , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...