Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Public Health ; 68: 1605459, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37711159

RESUMEN

Objectives: To assess health-related quality of life (QoL) in caregivers of elderly patients with chronic disabilities receiving, or not receiving, social worker support. Methods: This multicenter open-label randomized study assigned caregivers to receive an information booklet, exclusively, or with social worker support. Caregivers completed Short Form-36 (SF-36) and Hospital Anxiety Depression Scale quarterly, and Zarit Burden Interview each semester, for 24 months. We reported caregiver QoL mean changes at 12 and 24 months (M12, M24). Longitudinal QoL analysis up to M24 used mixed models for repeated measures (MMRM). Results: Among the 179 caregivers randomized from 2015 to 2019, the SF-36 physical and mental component summary showed no significant changes at M12 and M24, in terms of neither anxiety nor burden. However, depression significantly increased (M12: 1.4 ± 4.0; M24: 1.7 ± 4.1) with significant adjusted mean increase using MMRM at M24: 3.4 [0.6-2.5] in the control group, exclusively. Conclusion: These findings call for better recognition of the social support to prevent caregiver QoL deterioration and alleviate their depression early in the course of the disease. Clinical Trial Registration: ClinicalTrials.gov, identifier NCT02626377.


Asunto(s)
Cuidadores , Calidad de Vida , Anciano , Humanos , Estudios Prospectivos , Pacientes , Ansiedad
2.
Stem Cells Transl Med ; 3(1): 91-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24311701

RESUMEN

Smooth muscle cells (SMCs) and endothelial cells (ECs) are typically derived separately, with low efficiencies, from human pluripotent stem cells (hPSCs). The concurrent generation of these cell types might lead to potential applications in regenerative medicine to model, elucidate, and eventually treat vascular diseases. Here we report a robust two-step protocol that can be used to simultaneously generate large numbers of functional SMCs and ECs from a common proliferative vascular progenitor population via a two-dimensional culture system. We show here that coculturing hPSCs with OP9 cells in media supplemented with vascular endothelial growth factor, basic fibroblast growth factor, and bone morphogenetic protein 4 yields a higher percentage of CD31(+)CD34(+) cells on day 8 of differentiation. Upon exposure to endothelial differentiation media and SM differentiation media, these vascular progenitors were able to differentiate and mature into functional endothelial cells and smooth muscle cells, respectively. Furthermore, we were able to expand the intermediate population more than a billion fold to generate sufficient numbers of ECs and SMCs in parallel for potential therapeutic transplantations.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Endoteliales/citología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Células Madre Pluripotentes/citología , Trasplante de Células Madre/métodos , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Técnicas de Cocultivo , Células Nutrientes , Humanos , Ratones , Neovascularización Fisiológica , Regeneración
3.
Proc Natl Acad Sci U S A ; 109(1): 215-20, 2012 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-22178754

RESUMEN

Marfan syndrome (MFS) is a heritable connective tissue disorder caused by mutations in the gene coding for FIBRILLIN-1 (FBN1), an extracellular matrix protein. MFS is inherited as an autosomal dominant trait and displays major manifestations in the ocular, skeletal, and cardiovascular systems. Here we report molecular and phenotypic profiles of skeletogenesis in tissues differentiated from human embryonic stem cells and induced pluripotent stem cells that carry a heritable mutation in FBN1. We demonstrate that, as a biological consequence of the activation of TGF-ß signaling, osteogenic differentiation of embryonic stem cells with a FBN1 mutation is inhibited; osteogenesis is rescued by inhibition of TGF-ß signaling. In contrast, chondrogenesis is not perturbated and occurs in a TGF-ß cell-autonomous fashion. Importantly, skeletal phenotypes observed in human embryonic stem cells carrying the monogenic FBN1 mutation (MFS cells) are faithfully phenocopied by cells differentiated from induced pluripotent-stem cells derived independently from MFS patient fibroblasts. Results indicate a unique phenotype uncovered by examination of mutant pluripotent stem cells and further demonstrate the faithful alignment of phenotypes in differentiated cells obtained from both human embryonic stem cells and induced pluripotent-stem cells, providing complementary and powerful tools to gain further insights into human molecular pathogenesis, especially of MFS.


Asunto(s)
Huesos/patología , Células Madre Embrionarias/patología , Células Madre Pluripotentes Inducidas/patología , Síndrome de Marfan/patología , Secuencia de Bases , Huesos/metabolismo , Diferenciación Celular , Condrogénesis , Células Madre Embrionarias/metabolismo , Fibrilina-1 , Fibrilinas , Humanos , Síndrome de Marfan/metabolismo , Proteínas de Microfilamentos/genética , Datos de Secuencia Molecular , Osteogénesis , Fenotipo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
4.
Biol Reprod ; 84(6): 1258-71, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21368299

RESUMEN

Identification of genes involved in trophoblast differentiation is of great interest in understanding cellular and molecular mechanisms involved in placental development and is relevant clinically to fetal development, fertility, and maternal health. Herein, we investigated differentiation of human embryonic stem cells (hESCs) down the trophoblast lineage by culture with bone morphogenetic protein 4 (BMP4) over a 10-day period. Within 2 days, the stemness markers POU5F1 and NANOG were markedly down-regulated, followed temporally by up-regulation of the CDX2, KRT7, HLA-G, ID2, CGA, and CGB trophoblast markers. To understand, on a global scale, changes in the transcriptome during the differentiation of hESCs down the trophoblast lineage, a large-scale microarray analysis was performed. Through whole-genome analysis, more than 3800 genes displayed statistically significant and 2-fold or greater changes in expression during the time course. Of those genes that showed the largest increases, many were involved in processes associated with trophoblast biology; however, novel genes were also identified. Some of them are hypothesized to be associated mainly with extracellular matrix remodeling (e.g., NID2) and cell migration and invasion (e.g., RAB25). Using Ingenuity pathways analysis software to identify signaling pathways involved in trophoblast differentiation or function, we discovered that many genes are involved in WNT/beta-catenin, ERK/MAPK, NFKB, and calcium signaling pathways, suggesting potential roles for these families in trophoblast development. This work provides an in vitro functional genomic model with which to identify genes involved in trophoblast development.


Asunto(s)
Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Trofoblastos/fisiología , Biomarcadores , Diferenciación Celular , Linaje de la Célula , Células Madre Embrionarias/citología , Perfilación de la Expresión Génica , Humanos , Trofoblastos/citología , Regulación hacia Arriba
5.
Int J Dev Biol ; 53(4): 569-78, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19378248

RESUMEN

Embryonic stem (ES) cells which constitutively express the Pdx-1, Ngn-3, NeuroD1, Nkx2.2, and Nkx6.1 transcription factors were engineered by means of lentiviral vectors, following a multi-step infection procedure to successively generate ES cell lines expressing one, two, and three factors, respectively. Each ES cell line was allowed to differentiate into nestin+/Isl-1+ endocrine precursors, then into more mature pancreatic cells, and subsequently analysed for expression of Glc, Ins, and Sst, markers of alpha, beta and delta cells, respectively. Each ES cell line generated displayed a unique pattern of gene expression. The ES cell line expressing NeuroD1 displayed vastly elevated levels of Glc, Ins-1, Ins-2 and Sst, and showed an increase in Pdx-1, Pax-4, Nkx6.1, Isl-1, Glut-2 and GK transcript levels. Furthermore, immunofluorescence analysis revealed that differentiation of NeuroD1-expressing ES cells in nestin+/Isl-1+ multilineage progenitors, followed by the formation of C-peptide+/insulin+ clusters, was accelerated. Together, these results indicate that stable expression of NeuroD1 in ES cells facilitates differentiation into endocrine and insulin-producing cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Linaje de la Célula , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Sistema Endocrino/metabolismo , Células Secretoras de Insulina/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Biomarcadores/metabolismo , Sistema Endocrino/citología , Sistema Endocrino/embriología , Regulación del Desarrollo de la Expresión Génica , Proteína Homeobox Nkx-2.2 , Insulina/biosíntesis , Células Secretoras de Insulina/citología , Ratones , Factores de Transcripción/metabolismo
6.
Cloning Stem Cells ; 9(3): 327-38, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17907943

RESUMEN

Embryonic stem cells (ES) can spontaneously activate a pancreatic differentiation program in vitro, although with low efficiency. The aim was to improve such process by using viral mediated gene transduction. In this study, we have examined the suitability of using viral vectors to express key transcriptional factors involved in pancreatic development. ES cell lines that constitutively express Pdx1, a homeodomain protein involved in both exocrine and endocrine pancreatic development and differentiation, were established using a lentiviral vector. These cells were additionally infected with an adenovirus expressing p48, a bHLH factor that is also crucial for pancreatic development and acinar differentiation. Quantitative RT-PCR analysis demonstrated an increase in the expression of exocrine genes, including those coding for both digestive enzymes and transcription factors. Immunocytochemical staining also revealed an increase in the number of amylase-expressing cell clusters. However, other important genes involved in acinar cell maturation (i.e., Mist1) were not modulated under these conditions, suggesting that the cells display features of immature exocrine cells or because of an uncoupled gene expression of the exocrine differentiation program. Importantly, this effect was selective for the acinar lineage as the expression of a large set of endocrine markers remained unchanged. Therefore, combined expression of key genes involved in pancreatic development may be a promising approach to generate mature pancreatic exocrine cells.


Asunto(s)
Células Madre Embrionarias/citología , Proteínas de Homeodominio/biosíntesis , Páncreas Exocrino/citología , Transactivadores/biosíntesis , Factores de Transcripción/biosíntesis , Adenoviridae/genética , Animales , Diferenciación Celular , Línea Celular , Linaje de la Célula , Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/genética , Lentivirus/genética , Ratones , Páncreas Exocrino/metabolismo , Transactivadores/genética , Factores de Transcripción/genética
7.
Stem Cells ; 24(3): 547-56, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16239321

RESUMEN

Using flow cytometry measurements combined with quantitative analysis of cell cycle kinetics, we show that rhesus monkey embryonic stem cells (ESCs) are characterized by an extremely rapid transit through the G1 phase, which accounts for 15% of the total cell cycle duration. Monkey ESCs exhibit a non-phasic expression of cyclin E, which is detected during all phases of the cell cycle, and do not growth-arrest in G1 after gamma-irradiation, reflecting the absence of a G1 checkpoint. Serum deprivation or pharmacological inhibition of mitogen-activated protein kinase kinase (MEK) did not result in any alteration in the cell cycle distribution, indicating that ESC growth does not rely on mitogenic signals transduced by the Ras/Raf/MEK pathway. Taken together, these data indicate that rhesus monkey ESCs, like their murine counterparts, exhibit unusual cell cycle features in which cell cycle control mechanisms operating during the G1 phase are reduced or absent.


Asunto(s)
Ciclina E/biosíntesis , Embrión de Mamíferos/citología , Células Madre/citología , Animales , Células Cultivadas , Embrión de Mamíferos/metabolismo , Fase G1/fisiología , Fase G1/efectos de la radiación , Rayos gamma , Regulación de la Expresión Génica/efectos de la radiación , Macaca mulatta , Transducción de Señal/fisiología , Transducción de Señal/efectos de la radiación , Especificidad de la Especie , Células Madre/metabolismo
8.
Stem Cells ; 23(4): 594-603, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15790780

RESUMEN

Multipotential stem cells can be selected from the bone marrow by plastic adhesion, expanded, and cultured. They are able to differentiate not only into multiple cell types, including cartilage, bone, adipose and fibrous tissues, and myelosupportive stroma, but also into mesodermal (endothelium), neuroectodermal, or endodermal (hepatocytes) lineages. Our goal was to characterize the multipotential capacities of human mesenchymal stem cells (hMSCs) and to evaluate their ability to differentiate into insulin-secreting cells in vitro. hMSCs were obtained from healthy donors, selected by plastic adhesion, and phenotyped by fluorescence-activated cell sorter and reverse transcription-polymerase chain reaction analysis before and after infection with adenoviruses coding for mouse IPF1, HLXB9, and FOXA2 transcription factors involved early in the endocrine developmental pathway. We found that native hMSCs have a pluripotent phenotype (OCT4 expression and high telomere length) and constitutively express NKX6-1 at a low level but lack all other transcription factors implicated in beta-cell differentiation. In all hMSCs, we detected mRNA of cytokeratin 18 and 19, epithelial markers present in pancreatic ductal cells, whereas proconvertase 1/3 mRNA expression was detected only in some hMSCs. Ectopic expression of IPF1, HLXB9, and FOXA2 with or without islet coculture or islet-conditioned medium results in insulin gene expression. In conclusion, our results demonstrated that in vitro human bone marrow stem cells are able to differentiate into insulin-expressing cells by a mechanism involving several transcription factors of the beta-cell developmental pathway when cultured in an appropriate microenvironment.


Asunto(s)
Células de la Médula Ósea/citología , Células Secretoras de Insulina/citología , Insulina/biosíntesis , Células Madre Mesenquimatosas/citología , Factores de Transcripción/biosíntesis , Adenoviridae/genética , Adipocitos/citología , Adipocitos/metabolismo , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Factor Nuclear 3-beta del Hepatocito/biosíntesis , Factor Nuclear 3-beta del Hepatocito/genética , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Insulina/genética , Células Secretoras de Insulina/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Telómero/metabolismo , Transactivadores/biosíntesis , Transactivadores/genética , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...