Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Stem Cells ; 35(1): 248-255, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27256706

RESUMEN

Mesenchymal stem cells (MSCs) have been proven to be therapeutically effective against atopic dermatitis (AD) in preclinical studies. However, the safety and efficacy of MSCs against AD have not yet been investigated in a clinical study. To establish the safety and efficacy of human umbilical cord blood-derived MSCs (hUCB-MSCs) in AD, 34 adult patients with moderate-to-severe AD were enrolled in two phase trials with a follow-up for 1 month and 3 months, respectively. Patients were randomly allocated to receive low dose (2.5 × 107 ) or high dose (5.0 × 107 ) of hUCB-MSCs subcutaneously. An Eczema Area and Severity Index (EASI) score, Investigator's Global Assessment (IGA) score, Severity Scoring for Atopic Dermatitis (SCORAD) score, adverse effect assessments, and serum biomarker levels were evaluated as end points. A single treatment of hUCB-MSCs resulted in dose-dependent improvements in AD manifestation. Fifty-five percent of patients in high dose hUCB-MSC-treated group showed a 50% reduction in the EASI score. The IGA score and SCORAD score decreased by 33% and 50%, respectively, in high dose-treated group. Particularly, the administration of high dose hUCB-MSCs reduced the pruritus score by 58%. The serum IgE levels and number of blood eosinophils were downregulated by the treatment. No serious adverse events occurred, and none of the patients discontinued the trial due to adverse events. This is the first report to demonstrate a marked improvement of AD features with cell therapeutics. These data suggest that the infusion of hUCB-MSCs might be an effective therapy for patients with moderate-to-severe AD. Stem Cells 2017;35:248-255.


Asunto(s)
Dermatitis Atópica/terapia , Células Madre Mesenquimatosas/citología , Cordón Umbilical/citología , Adulto , Biomarcadores/metabolismo , Demografía , Dermatitis Atópica/patología , Determinación de Punto Final , Femenino , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
2.
Growth Factors ; 27(6): 425-37, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19919531

RESUMEN

It has widely been reported that basic fibroblast growth factor (bFGF) promotes proliferation of human stem cells and contributes to the maintenance of their self-renewal capability through repeated replications. In contrast to embryonic stem cells (ESCs), the effects of growth factors on adult stem cells are poorly understood. In human umbilical cord blood-derived multipotent stem cells (hUCB-MSCs), bFGF is associated with an increased number of proliferating cells. Furthermore, expression levels of ESC markers were increased after treatment with bFGF. bFGF also increased the expression of FGFR, which in turn increased expression of insulin-like growth factor (IGFs). Since IGFs exert autocrine and paracrine effects on stem cells, bFGF-mediated release of IGFs from hUCB-MSCs might enhance FGFR1 and IGF1R expression in neighboring cells. These receptors could subsequently regulate the effects of bFGF and IGFs in adult stem cells. These results suggest that positive feedback regulation of bFGF and IGFs leads to proliferation of hUCB-MSCs.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica , Factor II del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células Madre Multipotentes/efectos de los fármacos , Adulto , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Femenino , Sangre Fetal , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/genética , Células Madre Multipotentes/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
3.
Biochem Biophys Res Commun ; 384(1): 120-5, 2009 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-19394308

RESUMEN

The OCT4A gene, a POU homeodomain transcription factor, has been shown to be expressed in embryonic stem cells (ESC) as well as hUCB-MSCs. In this study, the roles played by OCT4A in hUCB-MSCs were determined by stably inhibiting OCT4A with lenti-viral vector-based small hairpin RNA (shRNA). A decreased rate of cell proliferation was observed in OCT4-inhibited hUCB-MSCs. Down-regulation of CCNA2 expression in OCT4-inhibited hUCB-MSCs was confirmed by RT-PCR and real-time RT-PCR analysis in three genetically independent hUCB-MSC clones. Adipogenic differentiation was also suppressed in OCT4-inhibited hUCB-MSCs. The up-regulation of DTX1 and down-regulation of HDAC1, 2, and 4 expressions may be related to this differentiation deformity. The expression of other transcription factors, including SOX2, REX1 and c-MYC, was also affected by OCT4 inhibition in hUCB-MSCs. In conclusion, these finding suggest that OCT4A performs functionally conserved roles in hUCB-MSCs, making its expression biologically important for ex vivo culture of hUCB-MSCs.


Asunto(s)
Sangre Fetal/citología , Células Madre Multipotentes/citología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Adipogénesis/genética , Ciclo Celular/genética , Diferenciación Celular/genética , Proliferación Celular , Células Cultivadas , Expresión Génica , Humanos , Células Madre Multipotentes/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/antagonistas & inhibidores
4.
Cytotherapy ; 11(3): 289-98, 1 p following 298, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19308770

RESUMEN

BACKGROUND AIMS: Mesenchymal stromal cells (MSC) have been studied intensively in regenerative medicine. However, their therapeutic potential against tumor formation and cancer metastasis is still unclear. The effects of transplantation of MSCs in early-stage of carcinogenesis, should be evaluated. METHODS: MSC isolated from human umbilical cord blood (UCB) and adipose tissue (AD) were transplanted in a mouse cancer metastasis model. The effects of MSC on tumor growth and metastasis were analyzed. The effects of transplantation of MSC into the mouse model at very early stage carcinogenesis were also evaluated. RESULTS: Human MSC reduced lung metastasis and inhibited the growth of human breast cancer cells by inducing apoptosis. In addition, transplantation of both UCB and AD MSC into a cancer model with no detectable clinical symptoms did not appear to promote tumor growth or metastasis. CONCLUSIONS: We evaluated the effect of MSC derived from human UCB and AD tissue in a tumor model. Our findings may help to elucidate the interaction between cancer cells and MSC, as well as the application of MSC to clinical trials.


Asunto(s)
Neoplasias de la Mama/terapia , Neoplasias Pulmonares/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Tejido Adiposo/citología , Animales , Antígenos de Diferenciación/metabolismo , Apoptosis , Neoplasias de la Mama/patología , Línea Celular Tumoral , Técnicas de Cocultivo , Femenino , Sangre Fetal/citología , Humanos , Neoplasias Pulmonares/secundario , Células Madre Mesenquimatosas/citología , Ratones , Ratones SCID , Trasplante de Neoplasias , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Embarazo , Ensayo de Tumor de Célula Madre
5.
Sci Rep ; 5: 8020, 2015 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-25620445

RESUMEN

DNA methyltransferase (DNMT) inhibitors regulate target gene expression through epigenetic modifications, and these compounds have primarily been studied for cancer therapy or reprogramming. However, the effect of DNMT inhibitors on the immunomodulatory capacity of human mesenchymal stem cells (hMSCs) has not been investigated. In the present study, we treated hMSCs with 5-azacytidine (5-aza), a DNMT inhibitor, and confirmed that the inhibitory effects on mononuclear cell proliferation and cell migration toward activated T cells were increased. To identify the immunomodulatory factors stimulated through 5-aza treatment, we investigated the changes in promoter methylation patterns using methylation arrays and observed that the promoters of immunomodulatory factors, COX2 and PTGES, and migration-related factors, CXCR2 and CXCR4, were hypomethylated after 5-aza treatment. In addition, we observed that the COX2-PGE2 pathway is one of the main pathways for the enhanced immunosuppressive activity of hMSCs through 5-aza treatment. We also determined that the migration of hMSCs toward ligands for CXCR2/CXCR4 was increased after 5-aza treatment. Moreover, using an experimental colitis model, we showed that 5-aza pre-treatment could enhance the therapeutic effect of MSCs against immune-related diseases.


Asunto(s)
Ciclooxigenasa 2/biosíntesis , ADN (Citosina-5-)-Metiltransferasas/genética , Oxidorreductasas Intramoleculares/biosíntesis , Receptores CXCR4/biosíntesis , Receptores de Interleucina-8B/biosíntesis , Azacitidina/administración & dosificación , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Ciclooxigenasa 2/genética , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Metilación de ADN/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Inmunomodulación/efectos de los fármacos , Oxidorreductasas Intramoleculares/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Regiones Promotoras Genéticas , Prostaglandina-E Sintasas , Receptores CXCR4/genética , Receptores de Interleucina-8B/genética , Transducción de Señal/genética , Linfocitos T/inmunología
6.
Korean J Ophthalmol ; 17(2): 140-4, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14717493

RESUMEN

To clarify the diabetic complications mediated by increased platelet activity, we undertook a study of the mean platelet component (MPC), as determined by an automated hematologic analyzer (ADVIA 120, Bayer). Prothrombin time (PT), activated partial thromboplastin time (aPTT), and fibrinogen were also measured to investigate blood viscosity abnormalities. MPC was determined in 100 healthy controls and in 100 diabetic patients, the latter of which were subdivided into no diabetic retinopathy (DR) (n = 25), nonproliferative DR (n = 30), and proliferative DR (n = 45) groups. The mean MPC level was 26.9 g/dl in the control group and 22.5 g/dl in the diabetic patients (p < 0.05). PT and aPTT were similar for the diabetic patients and the controls; however, their corresponding fibrinogen levels were significantly different between the two groups(3.26 +/- 1.14 g/L vs. 4.21 +/- 2.35 g/L, p < 0.05). Our results suggest that platelet hyperfunction in diabetic patients may be implicated in the pathogenesis of diabetic retinopathy.


Asunto(s)
Retinopatía Diabética/sangre , Activación Plaquetaria , Adulto , Estudios de Casos y Controles , Femenino , Fibrinógeno/análisis , Humanos , Masculino , Persona de Mediana Edad , Tiempo de Tromboplastina Parcial , Tiempo de Protrombina
7.
Cell Transplant ; 22(1): 29-40, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22963690

RESUMEN

ZNF281 is one of the core transcription factors in embryonic stem cells (ESCs) and has activation and repression roles in the transcription of ESC genes. A known target molecule of Zfp281 (the mouse homologue of ZNF281) is Nanog. However, NANOG is not expressed in most human multipotent stem cells (hMSCs). Here, we investigated the roles of ZNF281 with a gain- and loss-of-function study. The knockdown of ZNF281 in vivo and in vitro resulted in spontaneous osteochondrogenic differentiation and reduced the proliferation of hMSCs, as determined by cell morphology and molecular markers. When ZNF281-knockdown hMSCs were subcutaneously implanted into mice along with ß-tricalcium phosphate (ß-TCP), many cells were converted into osteoblasts within 4 weeks. In contrast, the overexpression of ZNF281 in hMSCs resulted in accelerated proliferation. The expression pattern of ZNF281 correlated well with the expression of ß-CATENIN during differentiation and in the gain/loss-of-function study in hMSCs. The binding of ZNF281 to the promoter region of ß-CATENIN was observed using a chromatin immunoprecipitation (ChIP) assay. In conclusion, we propose that ZNF281 plays an important role in the maintenance and osteogenic differentiation of stem cells via the transcriptional regulation of genes including ß-CATENIN.


Asunto(s)
Osteocitos/fisiología , Transactivadores/deficiencia , Animales , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Trasplante de Células Madre de Sangre del Cordón Umbilical , Sangre Fetal/citología , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Desnudos , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Osteocitos/citología , Osteocitos/metabolismo , Osteogénesis/genética , Proteínas Represoras , Transactivadores/biosíntesis , Transactivadores/genética , Transfección
8.
PLoS One ; 5(5): e10493, 2010 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-20463961

RESUMEN

BACKGROUND: REX1/ZFP42 is a well-known embryonic stem cell (ESC) marker. However, the role of REX1, itself, is relatively unknown because the function of REX1 has only been reported in the differentiation of ESCs via STAT signaling pathways. Human mesenchymal stem cells (hMSCs) isolated from young tissues and cancer cells express REX1. METHODOLOGY/PRINCIPAL FINDING: Human umbilical cord blood-derived MSCs (hUCB-MSCs) and adipose tissue-derived MSCs (hAD-MSCs) strongly express REX1 and have a lower activation status of p38 MAPK, but bone marrow-derived MSCs (hBM-MSCs) have weak REX1 expression and higher activation of p38 MAPK. These results indicated that REX1 expression in hMSCs was positively correlated with proliferation rates but inversely correlated with the phosphorylation of p38 MAPK. In hUCB-MSCs, the roles of REX1 and p38 MAPK were investigated, and a knockdown study was performed using a lentiviral vector-based small hairpin RNA (shRNA). After REX1 knockdown, decreased cell proliferation was observed. In REX1 knocked-down hUCB-MSCs, the osteogenic differentiation ability deteriorated, but the adipogenic potential increased or was similar to that observed in the controls. The phosphorylation of p38 MAPK in hUCB-MSCs significantly increased after REX1 knockdown. After p38 MAPK inhibitor treatment, the cell growth in REX1 knocked-down hUCB-MSCs almost recovered, and the suppressed expression levels of CDK2 and CCND1 were also restored. The expression of MKK3, an upstream regulator of p38 MAPK, significantly increased in REX1 knocked-down hUCB-MSCs. The direct binding of REX1 to the MKK3 gene was confirmed by a chromatin immunoprecipitation (ChIP) assay. CONCLUSIONS/SIGNIFICANCE: These findings showed that REX1 regulates the proliferation/differentiation of hMSCs through the suppression of p38 MAPK signaling via the direct suppression of MKK3. Therefore, p38 MAPK and REX-1 status can determine the cell fate of adult stem cells (ASCs). These results were the first to show the role of REX1 in the proliferation/differentiation of ASCs.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Factores de Transcripción de Tipo Kruppel/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Adulto , Apoptosis , Proliferación Celular , Ciclinas/metabolismo , Activación Enzimática , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Factores de Transcripción de Tipo Kruppel/antagonistas & inhibidores , MAP Quinasa Quinasa 3/metabolismo , Fosforilación , Receptores Notch/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA