Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Int J Mol Sci ; 22(17)2021 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-34502036

RESUMEN

The success of cell therapy for the treatment of myocardial infarction depends on finding novel approaches that can substantially implement the engraftment of the transplanted cells. In order to enhance cell engraftment, most studies have focused on the pretreatment of transplantable cells. Here we have considered an alternative approach that involves the preconditioning of infarcted heart tissue to reduce endogenous cell activity and thus provide an advantage to our exogenous cells. This treatment is routinely used in other tissues such as bone marrow and skeletal muscle to improve cell engraftment, but it has never been taken in cardiac tissue. To avoid long-term cardiotoxicity induced by full heart irradiation we developed a rat model of a catheter-based heart irradiation system to locally impact a delimited region of the infarcted cardiac tissue. As proof of concept, we transferred ZsGreen+ iPSCs in the infarcted heart, due to their ease of use and detection. We found a very significant increase in cell engraftment in preirradiated rats. In this study, we demonstrate for the first time that preconditioning the infarcted cardiac tissue with local irradiation can substantially enhance cell engraftment.


Asunto(s)
Braquiterapia/métodos , Precondicionamiento Isquémico/métodos , Infarto del Miocardio/terapia , Trasplante de Células Madre/métodos , Animales , Células Cultivadas , Corazón/efectos de la radiación , Células Madre Pluripotentes Inducidas/trasplante , Masculino , Ratones , Ratas , Ratas Sprague-Dawley
2.
Int J Mol Sci ; 21(21)2020 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-33114756

RESUMEN

Direct cardiac reprogramming has emerged as a novel therapeutic approach to treat and regenerate injured hearts through the direct conversion of fibroblasts into cardiac cells. Most studies have focused on the reprogramming of fibroblasts into induced cardiomyocytes (iCMs). The first study in which this technology was described, showed that at least a combination of three transcription factors, GATA4, MEF2C and TBX5 (GMT cocktail), was required for the reprogramming into iCMs in vitro using mouse cells. However, this was later demonstrated to be insufficient for the reprogramming of human cells and additional factors were required. Thereafter, most studies have focused on implementing reprogramming efficiency and obtaining fully reprogrammed and functional iCMs, by the incorporation of other transcription factors, microRNAs or small molecules to the original GMT cocktail. In this respect, great advances have been made in recent years. However, there is still no consensus on which of these GMT-based varieties is best, and robust and highly reproducible protocols are still urgently required, especially in the case of human cells. On the other hand, apart from CMs, other cells such as endothelial and smooth muscle cells to form new blood vessels will be fundamental for the correct reconstruction of damaged cardiac tissue. With this aim, several studies have centered on the direct reprogramming of fibroblasts into induced cardiac progenitor cells (iCPCs) able to give rise to all myocardial cell lineages. Especially interesting are reports in which multipotent and highly expandable mouse iCPCs have been obtained, suggesting that clinically relevant amounts of these cells could be created. However, as of yet, this has not been achieved with human iCPCs, and exactly what stage of maturity is appropriate for a cell therapy product remains an open question. Nonetheless, the major concern in regenerative medicine is the poor retention, survival, and engraftment of transplanted cells in the cardiac tissue. To circumvent this issue, several cell pre-conditioning approaches are currently being explored. As an alternative to cell injection, in vivo reprogramming may face fewer barriers for its translation to the clinic. This approach has achieved better results in terms of efficiency and iCMs maturity in mouse models, indicating that the heart environment can favor this process. In this context, in recent years some studies have focused on the development of safer delivery systems such as Sendai virus, Adenovirus, chemical cocktails or nanoparticles. This article provides an in-depth review of the in vitro and in vivo cardiac reprograming technology used in mouse and human cells to obtain iCMs and iCPCs, and discusses what challenges still lie ahead and what hurdles are to be overcome before results from this field can be transferred to the clinical settings.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Factores de Transcripción/metabolismo , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Reprogramación Celular , Medios de Cultivo/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo
3.
Nature ; 465(7299): 808-12, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20535210

RESUMEN

The generation of reprogrammed induced pluripotent stem cells (iPSCs) from patients with defined genetic disorders holds the promise of increased understanding of the aetiologies of complex diseases and may also facilitate the development of novel therapeutic interventions. We have generated iPSCs from patients with LEOPARD syndrome (an acronym formed from its main features; that is, lentigines, electrocardiographic abnormalities, ocular hypertelorism, pulmonary valve stenosis, abnormal genitalia, retardation of growth and deafness), an autosomal-dominant developmental disorder belonging to a relatively prevalent class of inherited RAS-mitogen-activated protein kinase signalling diseases, which also includes Noonan syndrome, with pleomorphic effects on several tissues and organ systems. The patient-derived cells have a mutation in the PTPN11 gene, which encodes the SHP2 phosphatase. The iPSCs have been extensively characterized and produce multiple differentiated cell lineages. A major disease phenotype in patients with LEOPARD syndrome is hypertrophic cardiomyopathy. We show that in vitro-derived cardiomyocytes from LEOPARD syndrome iPSCs are larger, have a higher degree of sarcomeric organization and preferential localization of NFATC4 in the nucleus when compared with cardiomyocytes derived from human embryonic stem cells or wild-type iPSCs derived from a healthy brother of one of the LEOPARD syndrome patients. These features correlate with a potential hypertrophic state. We also provide molecular insights into signalling pathways that may promote the disease phenotype.


Asunto(s)
Células Madre Pluripotentes Inducidas/patología , Síndrome LEOPARD/patología , Modelos Biológicos , Medicina de Precisión , Adulto , Diferenciación Celular , Línea Celular , Linaje de la Célula , Células Cultivadas , Células Madre Embrionarias/metabolismo , Activación Enzimática , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Células Madre Pluripotentes Inducidas/enzimología , Células Madre Pluripotentes Inducidas/metabolismo , Síndrome LEOPARD/tratamiento farmacológico , Síndrome LEOPARD/metabolismo , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Fosfoproteínas/análisis , Reacción en Cadena de la Polimerasa , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Factores de Transcripción SOXB1/genética
4.
Stem Cell Reports ; 18(1): 64-80, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36563688

RESUMEN

Naive human pluripotent stem cells (hPSCs) are defined as the in vitro counterpart of the human preimplantation embryo's epiblast and are used as a model system to study developmental processes. In this study, we report the discovery and characterization of distinct cell populations coexisting with epiblast-like cells in 5iLAF naive human induced PSC (hiPSC) cultures. It is noteworthy that these populations closely resemble different cell types of the human embryo at early developmental stages. While epiblast-like cells represent the main cell population, interestingly we detect a cell population with gene and transposable element expression profile closely resembling the totipotent eight-cell (8C)-stage human embryo, and three cell populations analogous to trophectoderm cells at different stages of their maturation process: transition, early, and mature stages. Moreover, we reveal the presence of cells resembling primitive endoderm. Thus, 5iLAF naive hiPSC cultures provide an excellent opportunity to model the earliest events of human embryogenesis, from the 8C stage to the peri-implantation period.


Asunto(s)
Embrión de Mamíferos , Células Madre Pluripotentes , Humanos , Desarrollo Embrionario/genética , Endodermo , Estratos Germinativos , Diferenciación Celular/genética , Blastocisto
5.
Dev Cell ; 58(24): 2881-2895.e7, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-37967560

RESUMEN

Generating organs from stem cells through blastocyst complementation is a promising approach to meet the clinical need for transplants. In order to generate rejection-free organs, complementation of both parenchymal and vascular cells must be achieved, as endothelial cells play a key role in graft rejection. Here, we used a lineage-specific cell ablation system to produce mouse embryos unable to form both the cardiac and vascular systems. By mouse intraspecies blastocyst complementation, we rescued heart and vascular system development separately and in combination, obtaining complemented hearts with cardiomyocytes and endothelial cells of exogenous origin. Complemented chimeras were viable and reached adult stage, showing normal cardiac function and no signs of histopathological defects in the heart. Furthermore, we implemented the cell ablation system for rat-to-mouse blastocyst complementation, obtaining xenogeneic hearts whose cardiomyocytes were completely of rat origin. These results represent an advance in the experimentation towards the in vivo generation of transplantable organs.


Asunto(s)
Sistema Cardiovascular , Corazón , Células Madre Pluripotentes , Animales , Ratones , Ratas , Blastocisto , Células Endoteliales , Miocitos Cardíacos , Corazón/embriología , Sistema Cardiovascular/embriología
6.
Animals (Basel) ; 12(14)2022 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-35883376

RESUMEN

Each year, tens of thousands of people worldwide die of end-stage organ failure due to the limited availability of organs for use in transplantation. To meet this clinical demand, one of the last frontiers of regenerative medicine is the generation of humanized organs in pigs from pluripotent stem cells (PSCs) via blastocyst complementation. For this, organ-disabled pig models are needed. As endothelial cells (ECs) play a critical role in xenotransplantation rejection in every organ, we aimed to produce hematoendothelial-disabled pig embryos targeting the master transcription factor ETV2 via CRISPR-Cas9-mediated genome modification. In this study, we designed five different guide RNAs (gRNAs) against the DNA-binding domain of the porcine ETV2 gene, which were tested on porcine fibroblasts in vitro. Four out of five guides showed cleavage capacity and, subsequently, these four guides were microinjected individually as ribonucleoprotein complexes (RNPs) into one-cell-stage porcine embryos. Next, we combined the two gRNAs that showed the highest targeting efficiency and microinjected them at higher concentrations. Under these conditions, we significantly improved the rate of biallelic mutation. Hence, here, we describe an efficient one-step method for the generation of hematoendothelial-disabled pig embryos via CRISPR-Cas9 microinjection in zygotes. This model could be used in experimentation related to the in vivo generation of humanized organs.

7.
Curr Opin Cardiol ; 26(3): 223-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21451408

RESUMEN

PURPOSE OF REVIEW: The development of induced pluripotent stem cell (iPSC) technology has led to many advances in the areas of directed cell differentiation and characterization. New methods for generating iPSC-derived cardiomyocytes provide an invaluable resource for the study of certain cardiovascular disorders. This review highlights the current technology in this field, its application thus far to the study of genetic disorders of the RAS/MAPK pathway and long-QT syndrome (LQTS), and future directions for the field. RECENT FINDINGS: Enhanced methods increase the efficiency of generating and stringently purifying iPSC-derived cardiomyocytes. The use of cardiomyocytes derived from patients with LEOPARD syndrome and LQTS has shed light on the molecular mechanisms of disease and validated their use as reliable human disease models. SUMMARY: The use of iPSC-derived cardiomyocytes to study genetic cardiovascular disorders will enable a deeper and more applicable understanding of the molecular mechanisms of human disease, as well as improving our ability to achieve successful cell-based therapies. Methods to efficiently generate these cells are improving and provide promise for future applications of this technology.


Asunto(s)
Enfermedades Cardiovasculares/genética , Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Diferenciación Celular , Fibroblastos , Humanos , Síndrome de QT Prolongado/genética , Modelos Genéticos
8.
Front Cell Dev Biol ; 9: 797927, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35127713

RESUMEN

Direct cardiac reprogramming has emerged as an interesting approach for the treatment and regeneration of damaged hearts through the direct conversion of fibroblasts into cardiomyocytes or cardiovascular progenitors. However, in studies with human cells, the lack of reporter fibroblasts has hindered the screening of factors and consequently, the development of robust direct cardiac reprogramming protocols.In this study, we have generated functional human NKX2.5GFP reporter cardiac fibroblasts. We first established a new NKX2.5GFP reporter human induced pluripotent stem cell (hiPSC) line using a CRISPR-Cas9-based knock-in approach in order to preserve function which could alter the biology of the cells. The reporter was found to faithfully track NKX2.5 expressing cells in differentiated NKX2.5GFP hiPSC and the potential of NKX2.5-GFP + cells to give rise to the expected cardiac lineages, including functional ventricular- and atrial-like cardiomyocytes, was demonstrated. Then NKX2.5GFP cardiac fibroblasts were obtained through directed differentiation, and these showed typical fibroblast-like morphology, a specific marker expression profile and, more importantly, functionality similar to patient-derived cardiac fibroblasts. The advantage of using this approach is that it offers an unlimited supply of cellular models for research in cardiac reprogramming, and since NKX2.5 is expressed not only in cardiomyocytes but also in cardiovascular precursors, the detection of both induced cell types would be possible. These reporter lines will be useful tools for human direct cardiac reprogramming research and progress in this field.

9.
Stem Cell Res ; 36: 101410, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30878013

RESUMEN

We have generated two human induced pluripotent stem cell (iPSC) lines from CD133+ cells isolated from umbilical cord blood (CB) of a female child using non-integrative Sendai virus. Here we describe the complete characterization of these iPSC lines: PRYDi-CB5 and PRYDi-CB40.


Asunto(s)
Antígeno AC133/genética , Línea Celular , Células Madre Pluripotentes Inducidas , Animales , Diferenciación Celular , Técnicas de Reprogramación Celular , Células Clonales , Sangre Fetal/citología , Marcadores Genéticos , Humanos , Cariotipo , Ratones Endogámicos NOD , Ratones SCID , Virus Sendai
10.
Cell Signal ; 19(4): 844-54, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17158029

RESUMEN

Interferon-alpha (IFN-alpha) has been used for the last 20 years in the maintenance therapy of multiple myeloma (MM), though it is only effective in some patients. Congruent with this, IFN-alpha induces apoptosis in some MM cell lines. Understanding the mechanism of IFN-alpha-induced apoptosis could be useful in establishing criteria of eligibility for therapy. Here we show that IFN-alpha-induced apoptosis in the MM cell lines U266 and H929 was completely blocked by a specific inhibitor of Jak1. The mTOR inhibitor rapamycin mitigated apoptosis in U266 but potentiated it in H929 cells. IFN-alpha induced PS exposure, DeltaPsi(m) loss and pro-apoptotic conformational changes of Bak, but not of Bax, and was fully prevented by Mcl-1 overexpression in U266 cells. IFN-alpha treatment caused the release of cytochrome c from mitochondria to cytosol and consequently, a limited proteolytic processing of caspases. Apoptosis induced by IFN-alpha was only slightly prevented by caspase inhibitors. Levels of the BH3-only proteins PUMA and Bim increased during IFN-alpha treatment. Bim increase and apoptosis was prevented by transfection with the siRNA for Bim. PUMA-siRNA transfection reduced electroporation-induced apoptosis but had no effect on apoptosis triggered by IFN-alpha. The potentiating effect of rapamycin on apoptosis in H929 cells was associated to an increase in basal and IFN-alpha-induced Bim levels. Our results indicate that IFN-alpha causes apoptosis in myeloma cells through a moderate triggering of the mitochondrial route initiated by Bim and that mTOR inhibitors may be useful in IFN-alpha maintenance therapy of certain MM patients.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Interferón-alfa/farmacología , Janus Quinasa 1/metabolismo , Proteínas de la Membrana/metabolismo , Mieloma Múltiple/patología , Proteínas Proto-Oncogénicas/metabolismo , Sirolimus/farmacología , Factor Inductor de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Caspasas/metabolismo , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citocromos c/metabolismo , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Glutatión/farmacología , Humanos , Janus Quinasa 1/antagonistas & inhibidores , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mieloma Múltiple/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos
11.
Cancer Res ; 66(11): 5781-9, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16740717

RESUMEN

Multiple myeloma represents an incurable disease, for which development of new therapies is required. Here, we report the effect on myeloma cells of LBH589, a new hydroxamic acid-derived histone deacetylase inhibitor. LBH589 was a potent antimyeloma agent (IC(50) < 40 nmol/L) on both cell lines and fresh cells from multiple myeloma patients, including cells resistant to conventional chemotherapeutic agents. In addition, LBH589 potentiated the action of drugs, such as bortezomib, dexamethasone, or melphalan. Using gene array, quantitative PCR, and Western analyses, we observed that LBH589 affected a large number of genes involved in cell cycle and cell death pathways. LBH589 blocked cell cycle progression, and this was accompanied by p21, p53, and p57 up-regulation. LBH589 induced cell death through an increase in the mitochondrial outer membrane permeability. LBH589 favored apoptosome formation by inducing cytochrome c release, Apaf-1 up-regulation, and caspase-9 cleavage. In addition, LBH589 stimulated a caspase-independent pathway through the release of AIF from the mitochondria. LBH589 down-regulated Bcl-2 and particularly Bcl-X. Moreover, overexpression of Bcl-X in multiple myeloma cells prevented LBH589-induced cell death. All these data indicate that LBH589 could be a useful drug for the treatment of multiple myeloma patients.


Asunto(s)
Inhibidores de Histona Desacetilasas , Mieloma Múltiple/tratamiento farmacológico , Acetilación/efectos de los fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/farmacología , Bortezomib , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Dexametasona/administración & dosificación , Dexametasona/farmacología , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/farmacología , Indoles , Melfalán/administración & dosificación , Melfalán/farmacología , Mieloma Múltiple/enzimología , Mieloma Múltiple/patología , Panobinostat , Pirazinas/administración & dosificación , Pirazinas/farmacología , Proteína bcl-X/biosíntesis
12.
Stem Cell Res ; 33: 125-129, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30343102

RESUMEN

Islet-1 (Isl1) is a transcription factor essential for life expressed in specific cells with different developmental origins. We have generated iPSC lines from fibroblasts of the transgenic Ai6 x Isl1-Cre (Ai6IslCre) mouse. Here we describe the complete characterization of four iPSC lines: ATCi-Ai6IslCre10, ATCi-Ai6IslCre35, ATCi-Ai6IslCre74 and ATCi-Ai6IslCre80.


Asunto(s)
Proteínas con Homeodominio LIM/genética , Factores de Transcripción/genética , Animales , Diferenciación Celular , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Ratones
13.
Stem Cell Res ; 21: 1-4, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28677526

RESUMEN

We generated ATCi-MF1 induced pluripotent stem (iPS) cell line from Macaca fascicularis adult skin fibroblasts using non-integrative Sendai viruses carrying OCT3/4, KLF4, SOX2 and c-MYC. Once established, ATCi-MF1 cells present a normal karyotype, are Sendai virus-free and express pluripotency associated markers. Microsatellite markers analysis confirmed the origin of the iPS cells from the parental fibroblasts. Pluripotency was tested with the in vivo teratoma formation assay. ATCi-MF1 cell line may be a useful primate iPS cell model to test different experimental conditions where the use of human cells can imply ethical issues, as microinjection of pluripotent stem cells in pre-implantational embryos.


Asunto(s)
Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Virus Sendai , Piel/metabolismo , Factores de Transcripción , Transducción Genética , Animales , Línea Celular , Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , Factor 4 Similar a Kruppel , Macaca fascicularis , Piel/citología , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
14.
Stem Cell Res ; 16(3): 617-21, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27346195

RESUMEN

Mef2c Anterior Heart Field (AHF) enhancer is activated during embryonic heart development and it is expressed in multipotent cardiovascular progenitors (CVP) giving rise to endothelial and myocardial components of the outflow tract, right ventricle and ventricular septum. Here we have generated iPSC from transgenic Mef2c-AHF-Cre x Ai6(RCLZsGreen) mice. These iPSC will provide a novel tool to investigate the AHF-CVP and their cell progeny.


Asunto(s)
Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , Miocardio/citología , Animales , Diferenciación Celular , Reprogramación Celular , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Genotipo , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipo , Factores de Transcripción MEF2/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Teratoma/metabolismo , Teratoma/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Stem Cell Reports ; 7(4): 602-618, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27666791

RESUMEN

Induced pluripotent stem cells (iPSCs) are a powerful tool for disease modeling. They are routinely generated from healthy donors and patients from multiple cell types at different developmental stages. However, reprogramming leukemias is an extremely inefficient process. Few studies generated iPSCs from primary chronic myeloid leukemias, but iPSC generation from acute myeloid or lymphoid leukemias (ALL) has not been achieved. We attempted to generate iPSCs from different subtypes of B-ALL to address the developmental impact of leukemic fusion genes. OKSM(L)-expressing mono/polycistronic-, retroviral/lentiviral/episomal-, and Sendai virus vector-based reprogramming strategies failed to render iPSCs in vitro and in vivo. Addition of transcriptomic-epigenetic reprogramming "boosters" also failed to generate iPSCs from B cell blasts and B-ALL lines, and when iPSCs emerged they lacked leukemic fusion genes, demonstrating non-leukemic myeloid origin. Conversely, MLL-AF4-overexpressing hematopoietic stem cells/B progenitors were successfully reprogrammed, indicating that B cell origin and leukemic fusion gene were not reprogramming barriers. Global transcriptome/DNA methylome profiling suggested a developmental/differentiation refractoriness of MLL-rearranged B-ALL to reprogramming into pluripotency.


Asunto(s)
Transdiferenciación Celular/genética , Reprogramación Celular , Reordenamiento Génico , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Animales , Biomarcadores , Línea Celular Transformada , Línea Celular Tumoral , Análisis por Conglomerados , Metilación de ADN , Expresión Génica , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Xenoinjertos , Humanos , Ratones , Células Progenitoras Mieloides/metabolismo , Proteínas de Fusión Oncogénica/genética , Fenotipo , Células Precursoras de Linfocitos B/metabolismo , Transcriptoma , Translocación Genética
16.
Stem Cell Reports ; 3(6): 1118-31, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25458894

RESUMEN

Neurons obtained directly from human somatic cells hold great promise for disease modeling and drug screening. Available protocols rely on overexpression of transcription factors using integrative vectors and are often slow, complex, and inefficient. We report a fast and efficient approach for generating induced neural cells (iNCs) directly from human hematopoietic cells using Sendai virus. Upon SOX2 and c-MYC expression, CD133-positive cord blood cells rapidly adopt a neuroepithelial morphology and exhibit high expansion capacity. Under defined neurogenic culture conditions, they express mature neuronal markers and fire spontaneous action potentials that can be modulated with neurotransmitters. SOX2 and c-MYC are also sufficient to convert peripheral blood mononuclear cells into iNCs. However, the conversion process is less efficient and resulting iNCs have limited expansion capacity and electrophysiological activity upon differentiation. Our study demonstrates rapid and efficient generation of iNCs from hematopoietic cells while underscoring the impact of target cells on conversion efficiency.


Asunto(s)
Transdiferenciación Celular , Leucocitos Mononucleares/citología , Neuronas/citología , Antígeno AC133 , Antígenos CD/metabolismo , Proliferación Celular , Células Cultivadas , Senescencia Celular/genética , Sangre Fetal/citología , Expresión Génica , Perfilación de la Expresión Génica , Glicoproteínas/metabolismo , Humanos , Inmunofenotipificación , Leucocitos Mononucleares/metabolismo , Potenciales de la Membrana , Neuronas/metabolismo , Péptidos/metabolismo , Fenotipo
17.
Cardiovasc Res ; 95(3): 327-35, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22673369

RESUMEN

AIMS: A variety of human inherited heart diseases affect the normal functions of cardiomyocytes (CMs), endothelial cells (ECs), or smooth muscle cells (SMCs). To study human heart disease and generate cardiac cells for basic and translational research, an efficient strategy is needed for production of cardiac lineages from human stem cells. In the present study, a highly reproducible method was developed that can simultaneously enrich a large number of CMs and cardiac SMCs and ECs from human induced pluripotent stem (iPS) cells with high purity. METHODS AND RESULTS: Human multipotent cardiovascular progenitor cells were generated from human iPS cells, followed by selective differentiation of the multipotent cardiovascular progenitor cells into CMs, ECs, and SMCs. With further fluorescence-activated cell sorting, each of the three cardiovascular cell types could be enriched with high purity (>90%). These enriched cardiovascular cells exhibited specific gene expression signatures and normal functions when assayed both in vitro and in vivo. Moreover, CMs purified from iPS cells derived from a patient with LEOPARD syndrome, a disease characterized by cardiac hypertrophy, showed the expected up-regulated expression of genes associated with cardiac hypertrophy. CONCLUSIONS: Overall, our technical advance provides the means for generating a renewable resource of pure human cardiovascular cells that can be used to dissect the mechanisms of human inherited heart disease and for the future development of drug and cell therapeutics for heart disease.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Separación Celular , Células Madre Pluripotentes Inducidas/fisiología , Células Madre Multipotentes/fisiología , Miocitos Cardíacos/fisiología , Miocitos del Músculo Liso/fisiología , Animales , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Moléculas de Adhesión Celular Neuronal/metabolismo , Separación Celular/métodos , Células Cultivadas , Proteínas Fetales/metabolismo , Citometría de Flujo , Regulación de la Expresión Génica , Genotipo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Síndrome LEOPARD/genética , Síndrome LEOPARD/patología , Síndrome LEOPARD/fisiopatología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Multipotentes/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos del Músculo Liso/metabolismo , Fenotipo , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Factores de Tiempo
18.
Cell Stem Cell ; 11(2): 179-94, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22862944

RESUMEN

Many signals must be integrated to maintain self-renewal and pluripotency in embryonic stem cells (ESCs) and to enable induced pluripotent stem cell (iPSC) reprogramming. However, the exact molecular regulatory mechanisms remain elusive. To unravel the essential internal and external signals required for sustaining the ESC state, we conducted a short hairpin (sh) RNA screen of 104 ESC-associated phosphoregulators. Depletion of one such molecule, aurora kinase A (Aurka), resulted in compromised self-renewal and consequent differentiation. By integrating global gene expression and computational analyses, we discovered that loss of Aurka leads to upregulated p53 activity that triggers ESC differentiation. Specifically, Aurka regulates pluripotency through phosphorylation-mediated inhibition of p53-directed ectodermal and mesodermal gene expression. Phosphorylation of p53 not only impairs p53-induced ESC differentiation but also p53-mediated suppression of iPSC reprogramming. Our studies demonstrate an essential role for Aurka-p53 signaling in the regulation of self-renewal, differentiation, and somatic cell reprogramming.


Asunto(s)
Células Madre Embrionarias/metabolismo , Células Madre Pluripotentes/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Animales , Aurora Quinasa A , Aurora Quinasas , Diferenciación Celular , Línea Celular , Proliferación Celular , Células Madre Embrionarias/citología , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Fosforilación , Células Madre Pluripotentes/citología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Xenopus
19.
Stem Cell Investig ; 3: 59, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27868041
20.
J Pathol ; 208(1): 108-18, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16278822

RESUMEN

Malignant plasma cells in multiple myeloma home to the bone marrow (BM), accumulate in different niches and, in late disease, migrate from the BM into blood. These migratory events involve cell trafficking across extracellular matrix (ECM)-rich basement membranes and interstitial tissues. Metalloproteinases (MMP) degrade ECM and facilitate tumour cell invasion. The chemokine CXCL12 is expressed in the BM, and it was previously shown that it triggers myeloma cell migration and activation. In the present work we show that CXCL12 promotes myeloma cell invasion across Matrigel-reconstituted basement membranes and type I collagen gels. MMP-9 activity was required for invasion through Matrigel towards CXCL12, whereas TIMP-1, a MMP-9 inhibitor that we found to be expressed by myeloma and BM stromal cells, impaired the invasion. In addition, we show that the membrane-bound MT1-MMP metalloproteinase is expressed by myeloma cells and contributes to CXCL12-promoted myeloma cell invasion across Matrigel. Increase in MT1-MMP expression, as well as induction of its membrane polarization by CXCL12 in myeloma cells, might represent potential mechanisms contributing to this invasion. CXCL12-promoted invasion across type I collagen involved metalloproteinases different from MT1-MMP. These data indicate that CXCL12 could contribute to myeloma cell trafficking in the BM involving MMP-9 and MT1-MMP activities.


Asunto(s)
Membrana Basal/patología , Quimiocinas CXC/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Mieloma Múltiple/patología , Membrana Basal/metabolismo , Materiales Biocompatibles , Línea Celular Tumoral , Movimiento Celular/fisiología , Quimiocina CXCL12 , Colágeno , Colágeno Tipo I/metabolismo , Combinación de Medicamentos , Matriz Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Laminina , Metaloproteinasas de la Matriz/análisis , Metaloproteinasas de la Matriz Asociadas a la Membrana , Mieloma Múltiple/metabolismo , Invasividad Neoplásica , Proteínas de Neoplasias/metabolismo , Proteoglicanos , Células del Estroma/metabolismo , Células del Estroma/patología , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA