Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 343
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 162(2): 412-424, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26186193

RESUMEN

Induced pluripotency is a promising avenue for disease modeling and therapy, but the molecular principles underlying this process, particularly in human cells, remain poorly understood due to donor-to-donor variability and intercellular heterogeneity. Here, we constructed and characterized a clonal, inducible human reprogramming system that provides a reliable source of cells at any stage of the process. This system enabled integrative transcriptional and epigenomic analysis across the human reprogramming timeline at high resolution. We observed distinct waves of gene network activation, including the ordered re-activation of broad developmental regulators followed by early embryonic patterning genes and culminating in the emergence of a signature reminiscent of pre-implantation stages. Moreover, complementary functional analyses allowed us to identify and validate novel regulators of the reprogramming process. Altogether, this study sheds light on the molecular underpinnings of induced pluripotency in human cells and provides a robust cell platform for further studies. PAPERCLIP.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Epigénesis Genética , Perfilación de la Expresión Génica , Histona Demetilasas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo
2.
Cell ; 163(1): 230-45, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26365490

RESUMEN

Embryonic stem cells (ESCs) repress the expression of exogenous proviruses and endogenous retroviruses (ERVs). Here, we systematically dissected the cellular factors involved in provirus repression in embryonic carcinomas (ECs) and ESCs by a genome-wide siRNA screen. Histone chaperones (Chaf1a/b), sumoylation factors (Sumo2/Ube2i/Sae1/Uba2/Senp6), and chromatin modifiers (Trim28/Eset/Atf7ip) are key determinants that establish provirus silencing. RNA-seq analysis uncovered the roles of Chaf1a/b and sumoylation modifiers in the repression of ERVs. ChIP-seq analysis demonstrates direct recruitment of Chaf1a and Sumo2 to ERVs. Chaf1a reinforces transcriptional repression via its interaction with members of the NuRD complex (Kdm1a, Hdac1/2) and Eset, while Sumo2 orchestrates the provirus repressive function of the canonical Zfp809/Trim28/Eset machinery by sumoylation of Trim28. Our study reports a genome-wide atlas of functional nodes that mediate proviral silencing in ESCs and illuminates the comprehensive, interconnected, and multi-layered genetic and epigenetic mechanisms by which ESCs repress retroviruses within the genome.


Asunto(s)
Células Madre Embrionarias/virología , Retrovirus Endógenos/genética , Provirus/genética , Animales , Factor 1 de Ensamblaje de la Cromatina/genética , Factor 1 de Ensamblaje de la Cromatina/metabolismo , Células Madre de Carcinoma Embrionario/virología , Epigénesis Genética , Ratones , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo
3.
Cell ; 158(4): 903-915, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25126793

RESUMEN

Somatic cell reprogramming, directed differentiation of pluripotent stem cells, and direct conversions between differentiated cell lineages represent powerful approaches to engineer cells for research and regenerative medicine. We have developed CellNet, a network biology platform that more accurately assesses the fidelity of cellular engineering than existing methodologies and generates hypotheses for improving cell derivations. Analyzing expression data from 56 published reports, we found that cells derived via directed differentiation more closely resemble their in vivo counterparts than products of direct conversion, as reflected by the establishment of target cell-type gene regulatory networks (GRNs). Furthermore, we discovered that directly converted cells fail to adequately silence expression programs of the starting population and that the establishment of unintended GRNs is common to virtually every cellular engineering paradigm. CellNet provides a platform for quantifying how closely engineered cell populations resemble their target cell type and a rational strategy to guide enhanced cellular engineering.


Asunto(s)
Ingeniería Celular/métodos , Células Madre/citología , Biología de Sistemas/métodos , Animales , Redes Reguladoras de Genes , Humanos , Ratones
4.
Cell ; 158(4): 889-902, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25126792

RESUMEN

Engineering clinically relevant cells in vitro holds promise for regenerative medicine, but most protocols fail to faithfully recapitulate target cell properties. To address this, we developed CellNet, a network biology platform that determines whether engineered cells are equivalent to their target tissues, diagnoses aberrant gene regulatory networks, and prioritizes candidate transcriptional regulators to enhance engineered conversions. Using CellNet, we improved B cell to macrophage conversion, transcriptionally and functionally, by knocking down predicted B cell regulators. Analyzing conversion of fibroblasts to induced hepatocytes (iHeps), CellNet revealed an unexpected intestinal program regulated by the master regulator Cdx2. We observed long-term functional engraftment of mouse colon by iHeps, thereby establishing their broader potential as endoderm progenitors and demonstrating direct conversion of fibroblasts into intestinal epithelium. Our studies illustrate how CellNet can be employed to improve direct conversion and to uncover unappreciated properties of engineered cells.


Asunto(s)
Ingeniería Celular/métodos , Biología de Sistemas/métodos , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Ingeniería Celular/normas , Redes Reguladoras de Genes , Macrófagos/citología , Macrófagos/metabolismo , Ratones
5.
Cell ; 155(4): 778-92, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24209617

RESUMEN

Regeneration capacity declines with age, but why juvenile organisms show enhanced tissue repair remains unexplained. Lin28a, a highly conserved RNA-binding protein expressed during embryogenesis, plays roles in development, pluripotency, and metabolism. To determine whether Lin28a might influence tissue repair in adults, we engineered the reactivation of Lin28a expression in several models of tissue injury. Lin28a reactivation improved hair regrowth by promoting anagen in hair follicles and accelerated regrowth of cartilage, bone, and mesenchyme after ear and digit injuries. Lin28a inhibits let-7 microRNA biogenesis; however, let-7 repression was necessary but insufficient to enhance repair. Lin28a bound to and enhanced the translation of mRNAs for several metabolic enzymes, thereby increasing glycolysis and oxidative phosphorylation (OxPhos). Lin28a-mediated enhancement of tissue repair was negated by OxPhos inhibition, whereas a pharmacologically induced increase in OxPhos enhanced repair. Thus, Lin28a enhances tissue repair in some adult tissues by reprogramming cellular bioenergetics. PAPERCLIP:


Asunto(s)
Proteínas de Unión al ARN/metabolismo , Cicatrización de Heridas , Animales , Embrión de Mamíferos/metabolismo , Metabolismo Energético , Extremidades/fisiología , Folículo Piloso/fisiología , Humanos , Ratones , Ratones Transgénicos , MicroARNs/metabolismo , Regeneración
6.
Nature ; 606(7915): 747-753, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35705805

RESUMEN

Haematopoietic stem cells (HSCs) arise in the embryo from the arterial endothelium through a process known as the endothelial-to-haematopoietic transition (EHT)1-4. This process generates hundreds of blood progenitors, of which a fraction go on to become definitive HSCs. It is generally thought that most adult blood is derived from those HSCs, but to what extent other progenitors contribute to adult haematopoiesis is not known. Here we use in situ barcoding and classical fate mapping to assess the developmental and clonal origins of adult blood in mice. Our analysis uncovers an early wave of progenitor specification-independent of traditional HSCs-that begins soon after EHT. These embryonic multipotent progenitors (eMPPs) predominantly drive haematopoiesis in the young adult, have a decreasing yet lifelong contribution over time and are the predominant source of lymphoid output. Putative eMPPs are specified within intra-arterial haematopoietic clusters and represent one fate of the earliest haematopoietic progenitors. Altogether, our results reveal functional heterogeneity during the definitive wave that leads to distinct sources of adult blood.


Asunto(s)
Envejecimiento , Linaje de la Célula , Embrión de Mamíferos , Hematopoyesis , Células Madre Hematopoyéticas , Animales , Embrión de Mamíferos/citología , Células Madre Hematopoyéticas/citología , Ratones , Células Madre Multipotentes/citología
7.
Cell ; 151(6): 1151-4, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23217698

RESUMEN

The 2012 Nobel Prize in Medicine or Physiology recognizes the architects of two of the great paradigm-shifting discoveries of the last half-century of biology. In experiments performed nearly 50 years apart, Gurdon and Yamanaka made feasible the reawakening of pluripotency inherent in all cells and challenged forever our notions of cellular identity.


Asunto(s)
Premio Nobel , Fisiología/historia , Células Madre Pluripotentes/citología , Investigación con Células Madre , Animales , Reprogramación Celular , Historia del Siglo XIX , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Japón , Células Madre Pluripotentes/metabolismo , Reino Unido
8.
Cell ; 148(6): 1110-22, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22424223

RESUMEN

Although development leads unidirectionally toward more restricted cell fates, recent work in cellular reprogramming has proven that one cellular identity can strikingly convert into another, promising countless applications in biomedical research and paving the way for modeling diseases with patient-derived stem cells. To date, there has been little discussion of which disease models are likely to be most informative. Here, we review evidence demonstrating that, because environmental influences and epigenetic signatures are largely erased during reprogramming, patient-specific models of diseases with strong genetic bases and high penetrance are likely to prove most informative in the near term. We also discuss the implications of the new reprogramming paradigm in biomedicine and outline how reprogramming of cell identities is enhancing our understanding of cell differentiation and prospects for cellular therapies and in vivo regeneration.


Asunto(s)
Medicina Regenerativa , Trasplante de Células Madre , Técnicas de Cultivo de Célula , Reprogramación Celular , Enfermedad/genética , Epigenómica , Humanos , Células Madre Pluripotentes/citología , Células Madre/citología
9.
Cell ; 146(4): 607-20, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21854985

RESUMEN

Previous experiments suggest a connection between the N-alpha-acetylation of proteins and sensitivity of cells to apoptotic signals. Here, we describe a biochemical assay to detect the acetylation status of proteins and demonstrate that protein N-alpha-acetylation is regulated by the availability of acetyl-CoA. Because the antiapoptotic protein Bcl-xL is known to influence mitochondrial metabolism, we reasoned that Bcl-xL may provide a link between protein N-alpha-acetylation and apoptosis. Indeed, Bcl-xL overexpression leads to a reduction in levels of acetyl-CoA and N-alpha-acetylated proteins in the cell. This effect is independent of Bax and Bak, the known binding partners of Bcl-xL. Increasing cellular levels of acetyl-CoA by addition of acetate or citrate restores protein N-alpha-acetylation in Bcl-xL-expressing cells and confers sensitivity to apoptotic stimuli. We propose that acetyl-CoA serves as a signaling molecule that couples apoptotic sensitivity to metabolism by regulating protein N-alpha-acetylation.


Asunto(s)
Supervivencia Celular , Proteínas/metabolismo , Proteína bcl-X/metabolismo , Acetilación , Animales , Apoptosis , Caspasa 2/metabolismo , Línea Celular , Embrión de Mamíferos/citología , Técnicas de Inactivación de Genes , Células HeLa , Humanos , Células Jurkat , Ratones , Procesamiento Proteico-Postraduccional
10.
Cell ; 147(1): 81-94, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21962509

RESUMEN

The let-7 tumor suppressor microRNAs are known for their regulation of oncogenes, while the RNA-binding proteins Lin28a/b promote malignancy by inhibiting let-7 biogenesis. We have uncovered unexpected roles for the Lin28/let-7 pathway in regulating metabolism. When overexpressed in mice, both Lin28a and LIN28B promote an insulin-sensitized state that resists high-fat-diet induced diabetes. Conversely, muscle-specific loss of Lin28a or overexpression of let-7 results in insulin resistance and impaired glucose tolerance. These phenomena occur, in part, through the let-7-mediated repression of multiple components of the insulin-PI3K-mTOR pathway, including IGF1R, INSR, and IRS2. In addition, the mTOR inhibitor, rapamycin, abrogates Lin28a-mediated insulin sensitivity and enhanced glucose uptake. Moreover, let-7 targets are enriched for genes containing SNPs associated with type 2 diabetes and control of fasting glucose in human genome-wide association studies. These data establish the Lin28/let-7 pathway as a central regulator of mammalian glucose metabolism.


Asunto(s)
Glucosa/metabolismo , MicroARNs/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Resistencia a la Insulina , Ratones , Ratones Noqueados , Ratones Transgénicos , MicroARNs/genética , Obesidad/genética , Obesidad/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
11.
Cell ; 147(3): 577-89, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-22036566

RESUMEN

BMP and Wnt signaling pathways control essential cellular responses through activation of the transcription factors SMAD (BMP) and TCF (Wnt). Here, we show that regeneration of hematopoietic lineages following acute injury depends on the activation of each of these signaling pathways to induce expression of key blood genes. Both SMAD1 and TCF7L2 co-occupy sites with master regulators adjacent to hematopoietic genes. In addition, both SMAD1 and TCF7L2 follow the binding of the predominant lineage regulator during differentiation from multipotent hematopoietic progenitor cells to erythroid cells. Furthermore, induction of the myeloid lineage regulator C/EBPα in erythroid cells shifts binding of SMAD1 to sites newly occupied by C/EBPα, whereas expression of the erythroid regulator GATA1 directs SMAD1 loss on nonerythroid targets. We conclude that the regenerative response mediated by BMP and Wnt signaling pathways is coupled with the lineage master regulators to control the gene programs defining cellular identity.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Hematopoyesis , Transducción de Señal , Vía de Señalización Wnt , Animales , Proteínas de Unión al ADN/metabolismo , Humanos , Regeneración , Proteína Smad1/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Pez Cebra
12.
Nat Rev Genet ; 20(7): 377-388, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30737492

RESUMEN

The derivation of induced pluripotent stem cells (iPSCs) over a decade ago sparked widespread enthusiasm for the development of new models of human disease, enhanced platforms for drug discovery and more widespread use of autologous cell-based therapy. Early studies using directed differentiation of iPSCs frequently uncovered cell-level phenotypes in monogenic diseases, but translation to tissue-level and organ-level diseases has required development of more complex, 3D, multicellular systems. Organoids and human-rodent chimaeras more accurately mirror the diverse cellular ecosystems of complex tissues and are being applied to iPSC disease models to recapitulate the pathobiology of a broad spectrum of human maladies, including infectious diseases, genetic disorders and cancer.


Asunto(s)
Enfermedades Transmisibles/terapia , Enfermedades Genéticas Congénitas/terapia , Células Madre Pluripotentes Inducidas/citología , Modelos Biológicos , Neoplasias/terapia , Ingeniería de Tejidos/métodos , Animales , Diferenciación Celular , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Quimera/genética , Quimera/inmunología , Enfermedades Transmisibles/genética , Enfermedades Transmisibles/inmunología , Enfermedades Transmisibles/patología , Descubrimiento de Drogas/métodos , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/inmunología , Enfermedades Genéticas Congénitas/patología , Terapia Genética/métodos , Humanos , Células Madre Pluripotentes Inducidas/inmunología , Células Madre Pluripotentes Inducidas/trasplante , Modelos Animales , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Organoides/citología , Organoides/efectos de los fármacos , Organoides/inmunología , Trasplante de Tejidos/métodos , Trasplante Heterólogo
13.
Nat Rev Mol Cell Biol ; 14(6): 357-68, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23673969

RESUMEN

Pluripotent stem cells constitute a platform to model disease and developmental processes and can potentially be used in regenerative medicine. However, not all pluripotent cell lines are equal in their capacity to differentiate into desired cell types in vitro. Genetic and epigenetic variations contribute to functional variability between cell lines and heterogeneity within clones. These genetic and epigenetic variations could 'lock' the pluripotency network resulting in residual pluripotent cells or alter the signalling response of developmental pathways leading to lineage bias. The molecular contributors to functional variability and heterogeneity in both embryonic stem (ES) cells and induced pluripotent stem (iPS) cells are only beginning to emerge, yet they are crucial to the future of the stem cell field.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/metabolismo , Epigénesis Genética/fisiología , Células Madre Pluripotentes Inducidas/metabolismo , Transducción de Señal/fisiología , Animales , Células Madre Embrionarias/citología , Humanos , Células Madre Pluripotentes Inducidas/citología
14.
Cell ; 140(4): 445-9, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20178735

RESUMEN

Lin28, a highly conserved RNA-binding protein, has emerged as a modulator of the processing of the let-7 microRNA. This role for Lin28 has important implications for our mechanistic understanding of pluripotency, the timing of development, and oncogenesis.


Asunto(s)
MicroARNs/genética , Procesamiento Proteico-Postraduccional , Proteínas de Unión al ARN/genética , Animales , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Neoplasias/metabolismo , Células Madre/metabolismo
16.
Nat Rev Mol Cell Biol ; 13(7): 471-6, 2012 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-22678486

RESUMEN

The increasing momentum of stem cell research continues, with the better characterization of induced pluripotent stem (iPS) cells, the conversion of differentiated cells into different cell types and the use of pluripotent stem cells to generate whole tissues, among other advances. Here, six experts in the field of stem cell research compare different stem cell models and highlight the importance of pursuing complementary experimental approaches for a better understanding of pluripotency and differentiation and an informed approach to medical applications.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Investigación con Células Madre , Células Madre/citología , Animales , Bioética , Diferenciación Celular , Humanos , Ratones , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos
17.
Cell ; 136(6): 1136-47, 2009 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-19303855

RESUMEN

Interactions between developmental signaling pathways govern the formation and function of stem cells. Prostaglandin (PG) E2 regulates vertebrate hematopoietic stem cells (HSC). Similarly, the Wnt signaling pathway controls HSC self-renewal and bone marrow repopulation. Here, we show that wnt reporter activity in zebrafish HSCs is responsive to PGE2 modulation, demonstrating a direct interaction in vivo. Inhibition of PGE2 synthesis blocked wnt-induced alterations in HSC formation. PGE2 modified the wnt signaling cascade at the level of beta-catenin degradation through cAMP/PKA-mediated stabilizing phosphorylation events. The PGE2/Wnt interaction regulated murine stem and progenitor populations in vitro in hematopoietic ES cell assays and in vivo following transplantation. The relationship between PGE2 and Wnt was also conserved during regeneration of other organ systems. Our work provides in vivo evidence that Wnt activation in stem cells requires PGE2, and suggests the PGE2/Wnt interaction is a master regulator of vertebrate regeneration and recovery.


Asunto(s)
Dinoprostona/metabolismo , Desarrollo Embrionario , Células Madre Hematopoyéticas/metabolismo , Proteínas Wnt/metabolismo , Pez Cebra/metabolismo , Animales , Proliferación Celular , Supervivencia Celular , Células Madre Embrionarias/metabolismo , Hígado/fisiología , Ratones , Regeneración , Transducción de Señal , Pez Cebra/embriología , beta Catenina/metabolismo
18.
Nature ; 553(7689): 506-510, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29342143

RESUMEN

All haematopoietic cell lineages that circulate in the blood of adult mammals derive from multipotent haematopoietic stem cells (HSCs). By contrast, in the blood of mammalian embryos, lineage-restricted progenitors arise first, independently of HSCs, which only emerge later in gestation. As best defined in the mouse, 'primitive' progenitors first appear in the yolk sac at 7.5 days post-coitum. Subsequently, erythroid-myeloid progenitors that express fetal haemoglobin, as well as fetal lymphoid progenitors, develop in the yolk sac and the embryo proper, but these cells lack HSC potential. Ultimately, 'definitive' HSCs with long-term, multilineage potential and the ability to engraft irradiated adults emerge at 10.5 days post-coitum from arterial endothelium in the aorta-gonad-mesonephros and other haemogenic vasculature. The molecular mechanisms of this reverse progression of haematopoietic ontogeny remain unexplained. We hypothesized that the definitive haematopoietic program might be actively repressed in early embryogenesis through epigenetic silencing, and that alleviating this repression would elicit multipotency in otherwise lineage-restricted haematopoietic progenitors. Here we show that reduced expression of the Polycomb group protein EZH1 enhances multi-lymphoid output from human pluripotent stem cells. In addition, Ezh1 deficiency in mouse embryos results in precocious emergence of functional definitive HSCs in vivo. Thus, we identify EZH1 as a repressor of haematopoietic multipotency in the early mammalian embryo.


Asunto(s)
Células Madre Embrionarias/citología , Silenciador del Gen , Hematopoyesis , Células Madre Hematopoyéticas/citología , Linfocitos/citología , Células Madre Multipotentes/citología , Complejo Represivo Polycomb 2/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Cromatina/genética , Cromatina/metabolismo , Desarrollo Embrionario , Femenino , Humanos , Linfocitos/metabolismo , Ratones , Células Madre Pluripotentes/citología , Complejo Represivo Polycomb 2/química , Complejo Represivo Polycomb 2/deficiencia , Complejo Represivo Polycomb 2/genética
19.
Cell ; 132(4): 544-8, 2008 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-18295571

RESUMEN

Resident pools of somatic stem cells in many organs are responsible for tissue maintenance and repair. The goal of regenerative medicine is to exploit these cells either by transplanting them from an exogenous source or by activating endogenous stem cells pharmacologically. For diseases caused by mutations in a single gene, the therapeutic goal is tissue replacement using stem cells engineered to correct the genetic defect. However, a number of technical hurdles must be overcome before therapies based on pluripotent human stem cells can enter the clinic.


Asunto(s)
Células Madre Pluripotentes/citología , Trasplante de Células Madre , Terapia Genética , Humanos , Medicina Regenerativa , Ingeniería de Tejidos
20.
Cell ; 134(5): 877-86, 2008 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-18691744

RESUMEN

Tissue culture of immortal cell strains from diseased patients is an invaluable resource for medical research but is largely limited to tumor cell lines or transformed derivatives of native tissues. Here we describe the generation of induced pluripotent stem (iPS) cells from patients with a variety of genetic diseases with either Mendelian or complex inheritance; these diseases include adenosine deaminase deficiency-related severe combined immunodeficiency (ADA-SCID), Shwachman-Bodian-Diamond syndrome (SBDS), Gaucher disease (GD) type III, Duchenne (DMD) and Becker muscular dystrophy (BMD), Parkinson disease (PD), Huntington disease (HD), juvenile-onset, type 1 diabetes mellitus (JDM), Down syndrome (DS)/trisomy 21, and the carrier state of Lesch-Nyhan syndrome. Such disease-specific stem cells offer an unprecedented opportunity to recapitulate both normal and pathologic human tissue formation in vitro, thereby enabling disease investigation and drug development.


Asunto(s)
Línea Celular , Enfermedades Genéticas Congénitas/patología , Células Madre Pluripotentes/citología , Células de la Médula Ósea/citología , Fibroblastos/citología , Humanos , Cariotipificación , Células Madre Mesenquimatosas/citología , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA