Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Stem Cells Dev ; 27(7): 456-465, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29415626

RESUMEN

Angiogenesis is a complicated and sequential process that plays an important role in different physiological processes. Mesenchymal stem cells (MSCs), which are pluripotent stem cells, are widely used for the treatment of ischemic and traumatic diseases, and exosomes derived from these cells can also promote angiogenesis. Therefore, we aimed to uncover mechanisms to improve MSC exosome-mediated angiogenesis. For this study, we isolated human adipose-derived MSCs (hAD-MSCs) and assessed differentiation ability and markers. Cells were divided into hypoxia-treated MSCs (H-MSCs) and normoxia-treated MSCs (N-MSC), and exosomes were extracted by ultrafiltration. Exosomes (100 µg/mL) from H-MSCs and N-MSCs were added to human umbilical vein endothelial cells (HUVECs). Exosome uptake and the ability of endothelial cells to form tubes were detected in real time. Protein samples were collected at different time points to detect the expression of inhibitors (Vash1) and enhancers (Angpt1 and Flk1) of angiogenesis; we also assessed their related signaling pathways. We found that exosomes from the hypoxia group were more easily taken up by HUVECs; furthermore, their angiogenesis stimulatory activity was also significantly enhanced compared to that with exosomes from the normoxia group. HUVECs exposed to exosomes from H-MSCs significantly upregulated angiogenesis-stimulating genes and deregulated angiogenesis-inhibitory genes. The expression of vascular endothelial growth factor (VEGF) and activation of the protein kinase A (PKA) signaling pathway in HUVECs were significantly increased by hypoxia-exposed exosomes. Moreover, a PKA inhibitor was shown to significantly suppress angiogenesis. Finally, we concluded that hypoxia-exposed exosomes derived from hAD-MSCs can improve angiogenesis by activating the PKA signaling pathway and promoting the expression of VEGF. These results could be used to uncover safe and effective treatments for traumatic diseases.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Exosomas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica/fisiología , Transducción de Señal/fisiología , Tejido Adiposo/citología , Diferenciación Celular/genética , Hipoxia de la Célula , Proliferación Celular/genética , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Exosomas/ultraestructura , Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica/genética , Transducción de Señal/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
J Immunol ; 194(9): 4298-308, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25825436

RESUMEN

Mesenchymal stem/stromal cells (MSCs) can influence the destiny of hematopoietic stem/progenitor cells (HSCs) and exert broadly immunomodulatory effects on immune cells. However, how MSCs regulate the differentiation of regulatory dendritic cells (regDCs) from HSCs remains incompletely understood. In this study, we show that mouse bone marrow-derived Sca-1(+)Lin(-)CD117(-) MSCs can drive HSCs to differentiate into a novel IFN regulatory factor (IRF)8-controlled regDC population (Sca(+) BM-MSC-driven DC [sBM-DCs]) when cocultured without exogenous cytokines. The Notch pathway plays a critical role in the generation of the sBM-DCs by controlling IRF8 expression in an RBP-J-dependent way. We observed a high level of H3K27me3 methylation and a low level of H3K4me3 methylation at the Irf8 promoter during sBM-DC induction. Importantly, infusion of sBM-DCs could alleviate colitis in mice with inflammatory bowel disease by inhibiting lymphocyte proliferation and increasing the numbers of CD4(+)CD25(+) regulatory T cells. Thus, these data infer a possible mechanism for the development of regDCs and further support the role of MSCs in treating immune disorders.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Factores Reguladores del Interferón/metabolismo , Células Madre Mesenquimatosas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Antígenos Ly/metabolismo , Diferenciación Celular , Citocinas/metabolismo , Células Dendríticas/citología , Modelos Animales de Enfermedad , Expresión Génica , Histonas/metabolismo , Inmunomodulación , Mediadores de Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Factores Reguladores del Interferón/genética , Proteínas de la Membrana/metabolismo , Ratones , Modelos Biológicos , Fenotipo , Proteínas Proto-Oncogénicas c-kit/metabolismo
4.
Stem Cell Rev Rep ; 10(2): 295-303, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24390934

RESUMEN

BACKGROUND AND PURPOSE: Intravascular injection of mesenchymal stem cells (MSCs) has been found to cause considerable vascular obstructions which may lead to serious outcomes, particularly after intra-arterial injection. However, the underlying mechanisms have been poorly understood. METHODS: In this study, we fractionated MSCs that had been cultured in monolayer for six passages into small (average diameter = 17.9 µm) and large (average diameter 30.4 µm) populations according to their sizes, and examined their vascular obstructions after intra-internal carotid artery injection in rats and mice in comparison with MSCs derived from 3D spheroids which were uniformly smaller in size (average diameter 12.6 µm). RESULTS: We found that 3D MSCs did not cause detectable infarct in the brain as evidenced by MRI scan and TTC stain, 2D MSCs in small size caused a microinfarct in one of five animals, which was co-localized to the area of entrapped MSCs (labeled with DiI), while 2D MSCs in large size caused much larger infarcts in all five animals, and substantial amounts of DiI-positive MSCs were found in the infarct. Meanwhile, corresponding neurological defects were observed in the animals with stroke. In consistence, injection of 2D MSCs (average diameter 26.5) caused a marked loss of cortical neurons and their axons in Thy1-GFP transgenic mice and the activation of microglia in CX3CR1-GFP transgenic mice in the area with MSC entrapment. CONCLUSIONS: Our results suggest that the size of MSCs is a significant cause of MSC caused vascular obstructions and stroke.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/fisiología , Accidente Cerebrovascular/etiología , Animales , Separación Celular , Tamaño de la Célula , Corteza Cerebral/patología , Humanos , Inyecciones Intraarteriales , Iris/irrigación sanguínea , Iris/patología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Ratas , Ratas Sprague-Dawley , Retina/patología , Vasos Retinianos/patología , Accidente Cerebrovascular/patología
5.
Stem Cells Dev ; 23(9): 978-89, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24341685

RESUMEN

The therapeutic effect of mesenchymal stem cells (MSCs) in tissue repair/regeneration is substantially dampened by the loss of primitive properties and poor engraftment to target organs. In this study, the multipotency and cell sizes of human MSCs, which had been expanded in monolayer culture for several passages, were dramatically restored after an episode of three-dimensional (3D) spheroid culture. Unlike MSCs derived from monolayer, which caused embolism and blindness, MSCs derived from 3D spheroids did not cause vascular obstructions, after intra-carotid artery infusion in rats. Importantly, intra-carotid infusion of 1 million 3D spheroid MSCs in rats 24 h after middle cerebral artery occlusion and reperfusion resulted in engraftment of the cells into the lesion and significant (over 70%) reduction of infarct size along with restoration of neurologic function. Moreover, the enhanced effect of spheroid MSCs was coincided with significantly increased differentiation of the MSCs into neurons and markedly increased number of endogenous glial fibrillary acidic protein-positive neural progenitors in the peri-infarct boundary zone. However, the similarly administered monolayer MSCs resulted in a modest functional improvement. Our results suggest that 3D MSCs, in combination with intra-carotid delivery, may represent a novel therapeutic approach of MSCs for stroke.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Esferoides Celulares/metabolismo , Accidente Cerebrovascular/terapia , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Xenoinjertos , Células Madre Mesenquimatosas/citología , Ratas , Ratas Sprague-Dawley , Esferoides Celulares/citología , Esferoides Celulares/trasplante , Accidente Cerebrovascular/metabolismo
6.
J Cell Biochem ; 114(6): 1374-84, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23239100

RESUMEN

The ERK-MAPK signaling pathway plays a pivotal role during mesenchymal stem cell (MSC) differentiation. Studies have demonstrated that ERK-MAPK promotes adipogenesis and osteogenesis through the phosphorylation of differentiation-associated transcription factors and that it is the only active signaling in all three lineages (adipogenic, chondrogenic, and osteogenic) during MSC differentiation. Recent studies pointed to the significant roles of microRNA-21 (miR-21) during several physiological and pathological processes, especially stem cell fate determination. The miR-21 expression pattern is also correlated with ERK-MAPK activity. Here, we found that miR-21 expression is elevated and associated with an increased differentiation potential in MSCs during adipogenesis and osteogenesis. The overexpression of miR-21 elevated the expression level of the differentiation-associated genes PPARγ and Cbfa-1 during MSC differentiation, whereas miR-21 knockdown reduced the expression level of both genes. The ERK-MAPK signaling pathway activity had an increasing tendency to respond to miR-21 upregulation and a decreasing tendency to respond to miR-21 down-regulation during the first 4 days of adipogenesis and osteogenesis. Our data indicate that miR-21 modulated ERK-MAPK signaling activity by repressing SPRY2 expression, a known regulator of the receptor tyrosine kinase (RTK) signaling pathway, to affect the duration and magnitude of ERK-MAPK activity. The ERK-MAPK signaling pathway was regulated by Sprouty2 (SPRY2) expression via a miR-21-mediated mechanism during MSC differentiation.


Asunto(s)
Adipogénesis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/metabolismo , Células Madre Mesenquimatosas/fisiología , MicroARNs/fisiología , Interferencia de ARN , Regiones no Traducidas 3' , Tejido Adiposo/citología , Adulto , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Secuencia de Bases , Sitios de Unión , Separación Celular , Células Cultivadas , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Proteínas de la Membrana/genética , Osteogénesis , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
7.
Exp Biol Med (Maywood) ; 237(9): 1110-6, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22956625

RESUMEN

Enhancer of zeste homolog 2 (EZH2) is crucially involved in epigenetic silencing by acting as a histone methyltransferase. Although EZH2 is overexpressed in many solid cancers, the role of EZH2 in B-cell acute lymphoblastic leukemia (B-ALL) remains largely unexplored. In a microarray experiment, we found that EZH2 was significantly upregulated in Nalm-6 cells and this was associated with the silencing of tumor suppressor genes p21, p53 and phosphatase and tensin homolog (PTEN). The abnormal expression of these genes was further confirmed by quantitative realtime polymerase chain reaction and Western blot analysis on Nalm-6 cells. Chromatin immunoprecipitation assay showed that EZH2 and H3K27me3 were both enriched in the promoter region of PTEN and p21 in Nalm-6 cells but not in normal B cells. Functional analysis showed that siRNA-mediated EZH2 knockdown led to decreased proliferation and increased apoptosis of Nalm-6 cells, accompanied by the reactivation of PTEN and p21 expression. Furthermore, we found that EZH2 inhibitor deazaneplanocin A promoted vincristine sulfate-induced apoptosis of Nalm-6 cells. Taken together, our data suggest that EZH2 is overexpressed in B-ALL and promotes the progression of B-ALL by directly mediating the inactivation of tumor suppressor genes p21 and PTEN, and could serve as a potential epigenetic target for B-ALL therapy.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Fosfohidrolasa PTEN/genética , Complejo Represivo Polycomb 2/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Apoptosis , Linfocitos B/metabolismo , Línea Celular Transformada , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Proteína Potenciadora del Homólogo Zeste 2 , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Fosfohidrolasa PTEN/metabolismo , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Complejo Represivo Polycomb 2/genética , Interferencia de ARN , ARN Interferente Pequeño , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba , Vincristina/farmacología
8.
J Immunol ; 189(3): 1182-92, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22753940

RESUMEN

Suppression of immune response by mesenchymal stem/stromal cells (MSCs) is well documented. However, their regulatory effects on immune cells, especially regulatory dendritic cells, are not fully understood. We have identified a novel Sca-1(+)Lin(-)CD117(-) MSC population isolated from mouse embryonic fibroblasts (MEF) that suppressed lymphocyte proliferation in vitro. Moreover, the Sca-1(+)Lin(-)CD117(-) MEF-MSCs induced hematopoietic stem/progenitor cells to differentiate into novel regulatory dendritic cells (DCs) (Sca-1(+)Lin(-)CD117(-) MEF-MSC-induced DCs) when cocultured in the absence of exogenous cytokines. Small interfering RNA silencing showed that Sca-1(+)Lin(-)CD117(-) MEF-MSCs induced the generation of Sca-1(+)Lin(-)CD117(-) MEF-MSC-induced DCs via IL-10-activated SOCS3, whose expression was regulated by the JAK-STAT pathway. We observed a high degree of H3K4me3 modification mediated by MLL1 and a relatively low degree of H3K27me3 modification regulated by SUZ12 on the promoter of SOCS3 during SOCS3 activation. Importantly, infusion of Sca-1(+)CD117(-)Lin(-) MEF-MSCs suppressed the inflammatory response by increasing DCs with a regulatory phenotype. Thus, our results shed new light on the role of MSCs in modulating regulatory DC production and support the clinical application of MSCs to reduce the inflammatory response in numerous disease states.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Interleucina-10/fisiología , Células Madre Mesenquimatosas/inmunología , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Línea Celular , Técnicas de Cocultivo , Células Madre Embrionarias/inmunología , Células Madre Embrionarias/metabolismo , Fibroblastos/inmunología , Fibroblastos/metabolismo , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Sistema de Señalización de MAP Quinasas/inmunología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Células del Estroma/inmunología , Células del Estroma/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Regulación hacia Arriba/inmunología
9.
Exp Hematol ; 40(9): 761-70, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22634392

RESUMEN

Recent findings indicate that mesenchymal stem cells (MSCs) may act as a regulator of Th17 cell differentiation, however, the underlying mechanism is still under debate. To investigate the underlying mechanisms of MSCs' regulatory effect, mouse bone marrow-derived MSCs were cocultured with mouse CD4(+)CD25(low)CD44(low)CD62L(high) T cells in vitro, and the proportion of induced Th17 cells, cytokines secretion, and transcription factors expression were examined by flow cytometry, enzyme-linked immunosorbent assay, quantitative reverse transcription polymerase chain reaction, and Western blotting. For the first time, our results showed that bone marrow-derived MSCs were able to inhibit Th17 cell differentiation via interleukin (IL)-10 secretion as the Th17 cell proportion was significantly regained when IL-10 was neutralized, or expression of IL-10 by bone marrow-derived MSCs was downregulated by RNA interference technique. Furthermore, IL-10 may suppress expression of Rorγt, the key transcription factor for Th17 cells, both by activating suppressor of cytokine signaling 3 through signal transducers and activators of transcription 5 phosphorylation, and decreasing signal transducers and activators of transcription 3 binding, which is at the promoter of Rorγt. Thus, our results demonstrate the inhibitory effect of MSCs on Th17 cells differentiation, and suggest increased IL-10 secretion might be the key factor.


Asunto(s)
Diferenciación Celular , Interleucina-10/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Th17/metabolismo , Animales , Western Blotting , Células Cultivadas , Técnicas de Cocultivo , Citometría de Flujo , Interleucina-10/genética , Ratones , Ratones Endogámicos BALB C , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
10.
Stem Cell Rev Rep ; 8(1): 243-50, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21706142

RESUMEN

A growing body of preclinical evidence suggests that mesenchymal stem cells (MSCs) are effective for the structural and functional recovery of the infracted heart. Accordingly, clinical trials are underway to determine the benefit of MSC-based therapies. While systemic administration of MSCs is an attractive strategy, and is the route currently used for the administration of MSCs in clinical studies for myocardial infarction, the majority of infused cells do not appear to localize to infracted myocardium in animal studies. Recently, important progress has been made in identifying chemokine receptors critical for the migration and homing of MSCs. Here, we review recent literature regarding mechanisms of MSC homing and recruitment to the ischemic myocardium, and discuss potential influences of low engraftment rates of systemically administered MSCs to the infracted heart tissue on the effects of MSC-based therapies on myocardial infarction.


Asunto(s)
Quimiocinas/fisiología , Quimiotaxis , Células Madre Mesenquimatosas/fisiología , Miocardio/citología , Animales , Células de la Médula Ósea/citología , Quimiocina CXCL12/metabolismo , Humanos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/terapia , Miocardio/metabolismo , Receptores CXCR4/metabolismo , Receptores de Quimiocina/metabolismo , Receptores de Quimiocina/fisiología
11.
PLoS One ; 6(6): e20526, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21694780

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) hold great promise for the treatment of difficult diseases. As MSCs represent a rare cell population, ex vivo expansion of MSCs is indispensable to obtain sufficient amounts of cells for therapies and tissue engineering. However, spontaneous differentiation and aging of MSCs occur during expansion and the molecular mechanisms involved have been poorly understood. METHODOLOGY/PRINCIPAL FINDINGS: Human MSCs in early and late passages were examined for their expression of genes involved in osteogenesis to determine their spontaneous differentiation towards osteoblasts in vitro, and of genes involved in self-renewal and proliferation for multipotent differentiation potential. In parallel, promoter DNA methylation and hostone H3 acetylation levels were determined. We found that MSCs underwent aging and spontaneous osteogenic differentiation upon regular culture expansion, with progressive downregulation of TERT and upregulation of osteogenic genes such as Runx2 and ALP. Meanwhile, the expression of genes associated with stem cell self-renewal such as Oct4 and Sox2 declined markedly. Notably, the altered expression of these genes were closely associated with epigenetic dysregulation of histone H3 acetylation in K9 and K14, but not with methylation of CpG islands in the promoter regions of most of these genes. bFGF promoted MSC proliferation and suppressed its spontaneous osteogenic differentiation, with corresponding changes in histone H3 acetylation in TERT, Oct4, Sox2, Runx2 and ALP genes. CONCLUSIONS/SIGNIFICANCE: Our results indicate that histone H3 acetylation, which can be modulated by extrinsic signals, plays a key role in regulating MSC aging and differentiation.


Asunto(s)
Diferenciación Celular/genética , Senescencia Celular/genética , Epigénesis Genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Acetilación/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Estrógenos/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Citometría de Flujo , Histonas/metabolismo , Humanos , Células Madre Multipotentes/citología , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/metabolismo , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo
12.
Exp Hematol ; 39(6): 608-16, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21288479

RESUMEN

MicroRNAs (miRNAs) are short non-coding RNAs involved in post-trascriptional regulation of gene expression and diverse biological activities. They are crucial for self-renewal and behavior of embryonic stem cells, but their role in mesenchymal stem cells has been poorly understood. Recently emerging evidence suggests that miRNAs are closely involved in controlling key steps of mesenchymal stem cell differentiation into certain cell lineages. This review focuses on miRNAs identified recently that regulate mesenchymal stem cell differentiation and other activities.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/citología , MicroARNs/fisiología , Animales , Linaje de la Célula , Senescencia Celular , Humanos , Cicatrización de Heridas
13.
Exp Hematol ; 39(2): 214-224.e1, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21078360

RESUMEN

OBJECTIVE: Cyclosporine A (CsA), known as an effective immunosuppressive agent, is widely used in clinical fields. Mesenchymal stem cells may exert immunomodulatory effects on the immune system, but the exact mechanisms underlying them remain controversial. Here we investigated whether human adipose tissue-derived mesenchymal stem cells (AMSCs) facilitate in vitro the immunomodulatory effects of CsA and we explored the molecule mechanisms that may be involved. MATERIALS AND METHODS: Proliferation of T lymphocytes was measured by uptake of (3)H-thymidine. Transcription and production of interleukin-2 and interferon-γ were evaluated by real-time quantitative polymerase chain reaction, reverse transcription polymerase chain reaction, and enzyme-linked immunosorbent assay. Nuclear factor-κB (NF-κB) was assayed by Western blotting and electrophoretic mobility shift assay. Expression of Jagged-1, Jagged-2, and Delta-1 of AMSCs were surveyed by flow cytometric analysis and Western blotting. RESULTS: The combination of moderate-dose AMSCs and low-dose CsA was significantly more powerful than moderate-dose AMSCs or large-dose CsA alone in suppressing transcription and production of interleukin-2 and interferon-γ, activation of NF-κB, and proliferation of T lymphocytes. In addition, AMSCs expressed a high level of Jagged-1, which induced activation of Notch signaling in T lymphocytes, thus reducing NF-κB activity. Anti-Jagged-1 neutralizing antibody and N [N-(3, 5-difluorophenacetyl-L-alanyl)]-S-phenylglycine t-butyl ester could reverse this trend. CONCLUSIONS: Human AMSCs facilitate the immunosuppressive effect of CsA on T lymphocytes through Jagged-1/Notch-related inhibition of NF-κB signaling. The combination of AMSCs and CsA represents a rationale therapeutic approach aimed to prevent adverse effects of CsA while maintaining its adequate immunosuppressive effect. Expression of Jagged-1 on AMSCs may provide an effective mechanism for the immunomodulatory activity of AMSCs via direct cell-cell interaction.


Asunto(s)
Tejido Adiposo/citología , Proteínas de Unión al Calcio/metabolismo , Ciclosporina/farmacología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Células Madre Mesenquimatosas/metabolismo , FN-kappa B/fisiología , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Inmunosupresores/farmacología , Proteína Jagged-1 , Unión Proteica , Proteínas Serrate-Jagged , Linfocitos T/citología , Linfocitos T/metabolismo
14.
Stem Cells ; 28(5): 905-15, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20474078

RESUMEN

Our understanding of the role of bone marrow (BM)-derived cells in cutaneous homeostasis and wound healing had long been limited to the contribution of inflammatory cells. Recent studies, however, suggest that the BM contributes a significant proportion of noninflammatory cells to the skin, which are present primarily in the dermis in fibroblast-like morphology and in the epidermis in a keratinocyte phenotype; and the number of these BM-derived cells increases markedly after wounding. More recently, several studies indicate that mesenchymal stem cells derived from the BM could significantly impact wound healing in diabetic and nondiabetic animals, through cell differentiation and the release of paracrine factors, implying a profound therapeutic potential. This review discusses the most recent understanding of the contribution of BM-derived noninflammatory cells to cutaneous homeostasis and wound healing.


Asunto(s)
Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea/métodos , Procedimientos Quirúrgicos Dermatologicos , Regeneración/fisiología , Piel/citología , Células Madre/fisiología , Cicatrización de Heridas/fisiología , Animales , Células de la Médula Ósea/citología , Trasplante de Médula Ósea/tendencias , Humanos , Piel/lesiones , Células Madre/citología
15.
Blood ; 113(1): 46-57, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18832657

RESUMEN

Mesenchymal stem cells (MSCs), in addition to their multilineage differentiation, exert immunomodulatory effects on immune cells, even dendritic cells (DCs). However, whether they influence the destiny of full mature DCs (maDCs) remains controversial. Here we report that MSCs vigorously promote proliferation of maDCs, significantly reduce their expression of Ia, CD11c, CD80, CD86, and CD40 while increasing CD11b expression. Interestingly, though these phenotypes clearly suggest their skew to immature status, bacterial lipopolysaccharide (LPS) stimulation could not reverse this trend. Moreover, high endocytosic capacity, low immunogenicity, and strong immunoregulatory function of MSC-treated maDCs (MSC-DCs) were also observed. Furthermore we found that MSCs, partly via cell-cell contact, drive maDCs to differentiate into a novel Jagged-2-dependent regulatory DC population and escape their apoptotic fate. These results further support the role of MSCs in preventing rejection in organ transplantation and treatment of autoimmune disease.


Asunto(s)
Comunicación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/metabolismo , Proteínas de la Membrana/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Animales , Apoptosis/inmunología , Antígeno B7-2/metabolismo , Antígeno CD11b/metabolismo , Antígeno CD11c/metabolismo , Antígenos CD40/metabolismo , Diferenciación Celular/inmunología , División Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Proteínas Fluorescentes Verdes/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Inmunofenotipificación , Inflamación/inmunología , Proteína Jagged-2 , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos
16.
Cell Res ; 18(4): 500-7, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18364678

RESUMEN

Human mesenchymal stem cells (hMSCs) can home to tumor sites and inhibit the growth of tumor cells. Little is known about the underlying molecular mechanisms that link hMSCs to the targeted inhibition of tumor cells. In this study, we investigated the effects of hMSCs on two human hepatoma cell lines (H7402 and HepG2) using an animal transplantation model, a co-culture system and conditioned media from hMSCs. Animal transplantation studies showed that the latent time for tumor formation was prolonged and that the tumor size was smaller when SCID mice were injected with H7402 cells and an equal number of Z3 hMSCs. When co-cultured with Z3 cells, H7402 cell proliferation decreased, apoptosis increased, and the expression of Bcl-2, c-Myc, proliferating cell nuclear antigen (PCNA) and survivin was downregulated. After treatment with conditioned media derived from Z3 hMSC cultures, H4702 cells showed decreased colony-forming ability and decreased proliferation. Immunoblot analysis showed that beta-catenin, Bcl-2, c-Myc, PCNA and survivin expression was downregulated in H7402 and HepG2 cells. Taken together, our findings demonstrate that hMSCs inhibit the malignant phenotypes of the H7402 and HepG2 human liver cancer cell lines, which include proliferation, colony-forming ability and oncogene expression both in vitro and in vivo. Furthermore, our studies provide evidence that the Wnt signaling pathway may have a role in hMSC-mediated targeting and tumor cell inhibition.


Asunto(s)
Carcinoma Hepatocelular/patología , Células Madre Mesenquimatosas/citología , Animales , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Humanos , Ratones , Ratones SCID , Trasplante de Neoplasias , Transducción de Señal , Proteínas Wnt/genética
17.
Trends Biotechnol ; 26(2): 70-6, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18191262

RESUMEN

The discovery of several types of small RNAs (sRNAs) has led to a steady increase in available RNA databases. Many of these sRNAs remain to be validated and functionally characterized. Recent advances in microRNA (miRNA)-expression profiling of different tissues, stages of development and physiological or pathological states are beginning to be explored using several technological approaches. In this review, these recent advances in miRNA microarray technology and their applications, particularly in basic research and clinical diagnosis, will be summarized and discussed. The methods for miRNA enrichment and probe design and labeling will also be discussed with an emphasis on evaluation of predicted miRNA sequences, analysis of miRNA expression and exploration of the potential roles of miRNA sequences in the regulation of stem cell differentiation and tissue- and time-specific profiling patterns of their target genes.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Marcación de Gen/métodos , MicroARNs/genética , MicroARNs/aislamiento & purificación , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Adulto , Humanos , MicroARNs/biosíntesis
18.
Methods ; 43(2): 123-30, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17889799

RESUMEN

Although a large number of small RNAs (sRNAs) have been discovered, it is very likely that the screens conducted so far have not reached saturation. Recently, many methods for predicting and identifying new sRNAs have been developed. However, it remains unclear what the total number of sRNAs within a genome is and how many types of sRNAs exist in plants and animals. In this article, combined methods of dynamic programming prediction, enrichment of sRNAs, and microarray analysis are developed to screen and evaluate a new class of sRNAs from introns of human, protein-encoding genes. The methods used by our laboratories to design capture probes and label enriched small RNAs are thoroughly described here. The microarray results show that our modified technologies are useful to enhance sensitivity and specificity of arrays, identify expression patterns within different cells, and discover differential expression of sRNAs during the differentiation process of bone marrow stem cells. Accordingly, the combination of computational prediction and microarray analysis may be a feasible and practical approach for profiling studies of both known and predicted small RNAs.


Asunto(s)
Perfilación de la Expresión Génica/métodos , MicroARNs/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Animales , Línea Celular , Humanos , MicroARNs/aislamiento & purificación , Peso Molecular , Hibridación de Ácido Nucleico
20.
Cell Res ; 17(3): 240-8, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17325691

RESUMEN

Mesenchymal stem cells (MSCs), derived from adult tissues, are multipotent progenitor cells, which hold great promise for regenerative medicine. Recent studies have shown that MSCs are immunosuppressive in vivo and in vitro in both animals and humans. However, the mechanisms that govern these immune modulatory functions of MSCs remain largely elusive. Some studies with bulk populations of MSCs indicated that soluble factors such as PGE2 and TGFbeta are important, while others support a role for cell-cell contact. In this study, we intended to clarify these issues by examining immunosuppressive effects of cloned MSCs. We derived MSC clones from mouse bone marrow and showed that the majority of these clones were able to differentiate into adipocytes and osteoblast-like cells. Importantly, cells from these clones exhibited strong inhibitory effects on TCR activation-induced T cell proliferation in vitro, and injection of a small number of these cells promoted the survival of allogeneic skin grafts in mice. Conditioned medium from MSC cultures showed some inhibitory effect on anti-CD3 induced lymphocyte proliferation independent of PGE2 and TGFbeta. In comparison, direct co-culture of MSCs with stimulated lymphocytes resulted in much stronger immunosuppressive effect. Interestingly, the suppression was bi-directional, as MSC proliferation was also reduced in the presence of lymphocytes. Taking together, our findings with cloned MSCs demonstrate that these cells exert their immunosuppressive effects through both soluble factor(s) and cell-cell contact, and that lymphocytes and MSCs are mutually inhibitory on their respective proliferation.


Asunto(s)
Células de la Médula Ósea/inmunología , Tolerancia Inmunológica , Células Madre Mesenquimatosas/inmunología , Animales , Comunicación Celular/inmunología , Proliferación Celular/efectos de los fármacos , Células Clonales , Técnicas de Cocultivo , Supervivencia de Injerto , Ionomicina/farmacología , Activación de Linfocitos , Linfocitos/citología , Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante de Piel/inmunología , Acetato de Tetradecanoilforbol/farmacología , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...