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










Base de datos
Intervalo de año de publicación
2.
Anticancer Drugs ; 34(5): 669-679, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36730426

RESUMEN

It was recently found that lncRNA PROX1 antisense RNA 1 (PROX1-AS1) manifested oncogenicity in a variety of malignancies. This work intended to investigate the molecular mechanisms of PROX1-AS1 in colorectal cancer (CRC) development and immune evasion. In this study, both PROX1-AS1 and PD-L1 expressions were lifted in CRC tissues and cells. PROX1-AS1 interference restrained CRC cell proliferation, migration, invasion, as well as CD8 + T-lymphocyte apoptosis, but increased the cytotoxicity and percentage of CD8 + T lymphocytes. The inhibitory effects of PROX1-AS1 inhibition on CRC progression and immune escape were positively related to PD-L1 suppression. PROX1-AS1 absorbed miR-520d to upregulate PD-L1 expression. PROX1-AS1 facilitated CRC progression and immune escape by targeting miR-520d. Furthermore, PROX1-AS1 deletion impaired CRC tumor growth in vivo . To sum up, this study affirmed that PROX1-AS1 could absorb miR-520d to upregulate PD-L1 in CRC, thereby promoting tumor progression and immune escape.


Asunto(s)
Neoplasias Colorrectales , MicroARNs , ARN Largo no Codificante , Humanos , MicroARNs/genética , ARN Largo no Codificante/genética , Línea Celular Tumoral , Antígeno B7-H1/metabolismo , Neoplasias Colorrectales/patología , Proliferación Celular/genética , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica
3.
Bioact Mater ; 24: 387-400, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36632503

RESUMEN

Bone marrow-derived mesenchymal stem cell (MSC) is one of the most actively studied cell types due to its regenerative potential and immunomodulatory properties. Conventional cell expansion methods using 2D tissue culture plates and 2.5D microcarriers in bioreactors can generate large cell numbers, but they compromise stem cell potency and lack mechanical preconditioning to prepare MSC for physiological loading expected in vivo. To overcome these challenges, in this work, we describe a 3D dynamic hydrogel using magneto-stimulation for direct MSC manufacturing to therapy. With our technology, we found that dynamic mechanical stimulation (DMS) enhanced matrix-integrin ß1 interactions which induced MSCs spreading and proliferation. In addition, DMS could modulate MSC biofunctions including directing MSC differentiation into specific lineages and boosting paracrine activities (e.g., growth factor secretion) through YAP nuclear localization and FAK-ERK pathway. With our magnetic hydrogel, complex procedures from MSC manufacturing to final clinical use, can be integrated into one single platform, and we believe this 'all-in-one' technology could offer a paradigm shift to existing standards in MSC therapy.

4.
World J Gastroenterol ; 28(31): 4328-4337, 2022 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-36159007

RESUMEN

BACKGROUND: Bile acids play an important role in the amelioration of type 2 diabetes following duodenal-jejunal bypass (DJB). Serum bile acids are elevated postoperatively. However, the clinical relevance is not known. Bile acids in the peripheral circulation reflect the amount of bile acids in the gut. Therefore, a further investigation of luminal bile acids following DJB is of great significance. AIM: To investigate changes of luminal bile acids following DJB. METHODS: Salicylhydroxamic acid (SHAM), DJB, and DJB with oral chenodeoxycholic acid (CDCA) supplementation were performed in a high-fat-diet/streptozotocin-induced diabetic rat model. Body weight, energy intake, oral glucose tolerance test, luminal bile acids, serum ceramides and intestinal ceramide synthesis were analyzed at week 12 postoperatively. RESULTS: Compared to SHAM, DJB achieved rapid and durable improvement in glucose tolerance and led to increased total luminal bile acid concentrations with preferentially increased proportion of farnesoid X receptor (FXR) - inhibitory bile acids within the common limb. Intestinal ceramide synthesis was repressed with decreased serum ceramides, and this phenomenon could be partially antagonized by luminal supplementation of FXR activating bile acid CDCA. CONCLUSION: DJB significantly changes luminal bile acid composition with increased proportion FXR-inhibitory bile acids and reduces serum ceramide levels. There observations suggest a novel mechanism of bile acids in metabolic regulation after DJB.


Asunto(s)
Ácidos y Sales Biliares , Diabetes Mellitus Tipo 2 , Animales , Glucemia/metabolismo , Ceramidas , Ácido Quenodesoxicólico/farmacología , Duodeno/metabolismo , Duodeno/cirugía , Glucosa , Yeyuno/metabolismo , Yeyuno/cirugía , Ratas , Salicilamidas , Estreptozocina
5.
Cytotherapy ; 24(5): 456-472, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35227601

RESUMEN

Therapies using mesenchymal stromal cells (MSCs) to treat immune and inflammatory conditions are now at an exciting stage of development, with many MSC-based products progressing to phase II and III clinical trials. However, a major bottleneck in the clinical translation of allogeneic MSC therapies is the variable immunomodulatory properties of MSC products due to differences in their tissue source, donor heterogeneity and processes involved in manufacturing and banking. This variable functionality of MSC products likely contributes to the substantial inconsistency observed in the clinical outcomes of phase III trials of MSC therapies; several trials have failed to reach the primary efficacy endpoint. In this review, we discuss various strategies to consistently maintain or enhance the immunomodulatory potency of MSCs during ex vivo expansion, which will enable the manufacture of allogeneic MSC banks that have high potency and low variability. Biophysical and biochemical priming strategies, the use of culture additives such as heparan sulfates, and genetic modification can substantially enhance the immunomodulatory properties of MSCs during in vitro expansion. Furthermore, robust donor screening, the use of biomarkers to select for potent MSC subpopulations, and rigorous quality testing to improve the release criteria for MSC banks have the potential to reduce batch-to-batch heterogeneity and enhance the clinical efficacy of the final MSC product. Machine learning approaches to develop predictive models of individual patient response can enable personalized therapies and potentially establish correlations between in vitro potency measurements and clinical outcomes in human trials.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Mesenquimatosas , Humanos , Inmunomodulación
6.
World J Stem Cells ; 13(12): 1826-1844, 2021 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-35069985

RESUMEN

Mesenchymal stem cells (MSCs) represent the most clinically used stem cells in regenerative medicine. However, due to the disadvantages with primary MSCs, such as limited cell proliferative capacity and rarity in the tissues leading to limited MSCs, gradual loss of differentiation during in vitro expansion reducing the efficacy of MSC application, and variation among donors increasing the uncertainty of MSC efficacy, the clinical application of MSCs has been greatly hampered. MSCs derived from human pluripotent stem cells (hPSC-MSCs) can circumvent these problems associated with primary MSCs. Due to the infinite self-renewal of hPSCs and their differentiation potential towards MSCs, hPSC-MSCs are emerging as an attractive alternative for regenerative medicine. This review summarizes the progress on derivation of MSCs from human pluripotent stem cells, disease modelling and drug screening using hPSC-MSCs, and various applications of hPSC-MSCs in regenerative medicine. In the end, the challenges and concerns with hPSC-MSC applications are also discussed.

7.
Stem Cell Reports ; 14(2): 210-225, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32004493

RESUMEN

The effects of ascorbate on adult cell fate specification remain largely unknown. Using our stepwise and chemically defined system to derive lateral mesoderm progenitors from human pluripotent stem cells (hPSCs), we found that ascorbate increased the expression of mesenchymal stromal cell (MSC) markers, purity of MSCs, the long-term self-renewal and osteochondrogenic capacity of hPSC-MSCs in vitro. Moreover, ascorbate promoted MSC specification in an iron-dependent fashion, but not in a redox-dependent manner. Further studies revealed that iron synergized with ascorbate to regulate hPSC-MSC histone methylation, promote their long-term self-renewal, and increase their osteochondrogenic capacity. We found that one of the histone demethylases affected by ascorbate, KDM4B, was necessary to promote the specification of hPSC-MSCs. This mechanistic understanding led to the metabolic optimization of hPSC-MSCs with an extended lifespan in vitro and the ability to fully repair cartilage defects upon transplantation in vivo. Our results highlight the importance of ascorbate and iron metabolism in adult human cell fate specification.


Asunto(s)
Ácido Ascórbico/farmacología , Huesos/citología , Autorrenovación de las Células/efectos de los fármacos , Hierro/farmacología , Células Madre Mesenquimatosas/citología , Activinas/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Cartílago/patología , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Mesodermo/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Línea Primitiva/citología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Proteínas Wnt/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
8.
Transl Cancer Res ; 8(4): 1540-1549, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35116897

RESUMEN

BACKGROUND: Let-7d has been reported to serve as a tumor suppressor in numerous cancers, however, the function in rectum adenocarcinoma has not been illuminated. In this study, we aimed to explore whether let-7d functions in rectum adenocarcinoma and its functional significance links to ATP binding cassette subfamily C member 2 (ABCC2). METHODS: The expression patterns of let-7d and ABCC2 were gained from TCGA database. Then, cell proliferation, invasion and migration assays were conducted to detect the influence on rectum adenocarcinoma cells behaviors after over-expression of let-7d. Subsequently, the potential target gene of let-7d was predicted and identified through bioinformatics prediction analysis and luciferase reporter assay. Analyses against prognostic value and independent predictor were acquired from Kaplan-Meier, Univariate and Multivariate analysis of Cox regression. Finally, con-transfection experiments were performed to investigate let-7d/ABCC2 pairs function on rectum adenocarcinoma cells after co-transfected with let-7d mimic and si-ABCC2. mRNA and protein levels were assessed by reverse transcription quantitative polymerase chain reaction (qRT-PCR) and western blot. RESULTS: The data from TCGA indicated that let-7d was down-regulated in rectum adenocarcinoma samples, whilst ABCC2 was showed a trend of high expression and its overexpression hinted to worse overall survival of rectum adenocarcinoma patients. Cells proliferation, invasion and migration properties were restrained after over-expression of let-7d in SW837 cells. Further investigations showed that over-expression of let-7d induced the inhibitory effect on SW837 cells proliferative, migrant and invasive capacities was augmented by silencing ABCC2. CONCLUSIONS: All results in this study indicated that up-regulation of let-7d could suppress SW837 cells growth, invasion and migration abilities by reducing ABCC2 expression, providing a new insight into molecular mechanism of let-7d/ABCC2 as a significant mediator for tumor progression and development of rectum adenocarcinoma.

9.
Nat Cell Biol ; 19(5): 412-414, 2017 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-28446816

RESUMEN

The metabolic transition from mitochondrial oxidative phosphorylation (OXPHOS) to glycolysis is critical for somatic reprogramming of induced pluripotent stem cells (iPSCs). SIRT2 has now been established as a previously unknown regulator of this metabolic transition during somatic reprogramming by controlling the acetylation status of glycolytic enzymes.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes , Acetilación , Glucólisis , Humanos , Células Madre Pluripotentes Inducidas
10.
Nat Commun ; 7: 13396, 2016 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-27869129

RESUMEN

Recent efforts have attempted to convert non-blood cells into hematopoietic stem cells (HSCs) with the goal of generating blood lineages de novo. Here we show that hematopoietic transcription factors Scl, Lmo2, Runx1 and Bmi1 can convert a developmentally distant lineage (fibroblasts) into 'induced hematopoietic progenitors' (iHPs). Functionally, iHPs generate acetylcholinesterase+ megakaryocytes and phagocytic myeloid cells in vitro and can also engraft immunodeficient mice, generating myeloerythoid and B-lymphoid cells for up to 4 months in vivo. Molecularly, iHPs transcriptionally resemble native Kit+ hematopoietic progenitors. Mechanistically, reprogramming factor Lmo2 implements a hematopoietic programme in fibroblasts by rapidly binding to and upregulating the Hhex and Gfi1 genes within days. Moreover the reprogramming transcription factors also require extracellular BMP and MEK signalling to cooperatively effectuate reprogramming. Thus, the transcription factors that orchestrate embryonic hematopoiesis can artificially reconstitute this programme in developmentally distant fibroblasts, converting them into engraftable blood progenitors.


Asunto(s)
Reprogramación Celular , Fibroblastos/fisiología , Células Madre Hematopoyéticas/fisiología , Factores de Transcripción/fisiología , Acetilcolinesterasa/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Quinasas MAP Reguladas por Señal Extracelular , Regulación de la Expresión Génica , Genómica , Humanos , Megacariocitos/fisiología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos , Células Mieloides/fisiología , Fagocitos/fisiología , Análisis por Matrices de Proteínas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Stem Cells ; 34(2): 277-87, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26676652

RESUMEN

In recent years, the highly conserved promyelocytic leukemia zinc finger (PLZF, also known as ZBTB16, ZNF145) has attracted attention as a multifunctional transcription factor involved in major biological processes during development. As a transcription factor, PLZF shows tight regulation in its cell-type-specific and stage-specific expression patterns. Emerging evidence shows that PLZF regulates the balance of self-renewal and differentiation in stem cells. However, the gene regulatory network of PLZF is only beginning to be understood. In this review, we discuss the diverse functions of PLZF, in particular its role in self-renewal versus differentiation of stem cells. We also discuss the current state of knowledge on the gene regulatory network of PLZF, in conjunction with its upstream factors, post-translational modifications and binding cofactors for multiprotein complexes. This review aims to provide the reader with an in-depth understanding of the molecular mechanisms underlying PLZF and the potential applications in tissue regeneration.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Regulación de la Expresión Génica/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Células Madre/metabolismo , Animales , Humanos , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Células Madre/citología
12.
Stem Cells Transl Med ; 3(5): 586-98, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24646489

RESUMEN

Induced pluripotent stem cells (iPSCs) derived from somatic cells of patients can be a good model for studying human diseases and for future therapeutic regenerative medicine. Current initiatives to establish human iPSC (hiPSC) banking face challenges in recruiting large numbers of donors with diverse diseased, genetic, and phenotypic representations. In this study, we describe the efficient derivation of transgene-free hiPSCs from human finger-prick blood. Finger-prick sample collection can be performed on a "do-it-yourself" basis by donors and sent to the hiPSC facility for reprogramming. We show that single-drop volumes of finger-prick samples are sufficient for performing cellular reprogramming, DNA sequencing, and blood serotyping in parallel. Our novel strategy has the potential to facilitate the development of large-scale hiPSC banking worldwide.


Asunto(s)
Bancos de Muestras Biológicas , Células Madre Pluripotentes Inducidas/citología , Manejo de Especímenes/métodos , Femenino , Dedos , Humanos , Masculino , Medicina Regenerativa/métodos
13.
Stem Cells Dev ; 22(2): 268-78, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22765508

RESUMEN

Mesenchymal stem cells (MSCs) represent one of the most promising stem cells for a number of degenerative conditions due to their multipotency, immunoprivileged properties, and easy expansion in vitro. However, the limited life span of primary MSCs during in vitro expansion greatly hampers their use in clinical applications and basic research. Immortalization of MSCs will overcome this problem and may provide a very useful tool with which to study MSC biology. Here we showed that silencing p53 expression with lentivirus-mediated small interfering RNA delayed the senescence by extended passage number, but was not sufficient to immortalize primary MSCs. However, combination of p53 knockdown and human telomerase reverse transcriptase (hTERT) overexpression was sufficient to immortalize MSCs. The effects of p53 knockdown and hTERT overexpression on MSCs, including proliferation, colony formation, and differentiation, were determined. The resultant immortal MSCs displayed similar surface antigen profile to primary MSCs and retained MSC differentiation potential. Gene expression profile showed high similarity between immortalized MSCs and primary MSCs. In addition, immortalization-associated genes were also identified. Our data suggested immortalization of MSCs related to upregulation of cell cycle regulator and DNA repair genes enabling them to bypass cell crisis and complete mitosis. This study provides a new cellular model for basic studies of MSCs and understanding of the molecular basis of MSC immortalization.


Asunto(s)
Transformación Celular Neoplásica , Regulación Enzimológica de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Telomerasa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Condrogénesis , Técnicas de Silenciamiento del Gen , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Ratones , Ratones Desnudos , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Telomerasa/genética , Factores de Tiempo , Transcriptoma , Proteína p53 Supresora de Tumor/genética
14.
Tissue Eng Part B Rev ; 19(3): 254-63, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23150948

RESUMEN

The development of the musculoskeletal system is a complex process that involves very precise control of bone formation and growth as well as remodeling during postnatal life. Although the understanding of the transcriptional mechanisms of osteogenesis has increased considerably, the molecular regulatory basis, especially the gene regulatory network of osteogenic differentiation, is still poorly understood. This review provides the reader with an overview of the key transcription factors that govern bone formation, highlighting their function and regulation linked to Runt-related transcription factor 2 (Runx2). Runx2 as the master transcription factor of osteoblast differentiation, Twist, Msh homeobox 2 (Msx2), and promyelocytic leukemia zinc-finger protein (PLZF) acting upstream of Runx2, Osterix (Osx) acting downstream of Runx2, and activating transcription factor 4 (ATF4) and zinc-finger protein 521 (ZFP521) acting as cofactors of Runx2 are discussed, and their relevance for tissue engineering is presented. References are provided for more in-depth personal study.


Asunto(s)
Desarrollo Óseo/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación de la Expresión Génica , Transcripción Genética , Animales , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Humanos , Osteogénesis/genética
15.
Stem Cells Dev ; 21(11): 1966-76, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22133004

RESUMEN

Adult mesenchymal stem cells (MSCs) are an attractive cell source for cartilage tissue engineering. In vitro predifferentiation of MSCs has been explored as a means to enhance MSC-based articular cartilage repair. However, there remain challenges to control and prevent the premature progression of MSC-derived chondrocytes to the hypertrophy. This study investigated the temporal effect of transforming growth factor (TGF)-ß and ß-catenin signaling co-activation during MSC chondrogenic differentiation and evaluated the influence of these predifferentiation conditions to subsequent phenotypic development of the cartilage. MSCs were differentiated in chondrogenic medium that contained either TGFß alone, TGFß with transient ß-catenin coactivation, or TGFß with continuous ß-catenin coactivation. After in vitro differentiation, the pellets were transplanted into SCID mice. Both coactivation protocols resulted in the enhancement of chondrogenic differentiation of MSCs. Compared with TGFß activation, transient coactivation of TGFß-induction with ß-catenin activation resulted in heightened hypertrophy and formed highly ossified tissues with marrow-like hematopoietic tissue in vivo. The continuous coactivation of the 2 signaling pathways, however, resulted in inhibition of progression to hypertrophy, marked by the suppression of type X collagen, Runx2, and alkaline phosphatase expression, and did not result in ossified tissue in vivo. Chondrocytes of the continuous co-activation samples secreted significantly more parathyroid hormone-related protein (PTHrP) and expressed cyclin D1. Our results suggest that temporal co-activation of the TGFß signaling pathway with ß-catenin can yield cartilage of different phenotype, represents a potential MSC predifferentiation protocol before clinical implantation, and has potential applications for the engineering of cartilage tissue.


Asunto(s)
Cartílago/fisiología , Condrogénesis , Células Madre Mesenquimatosas/citología , Fenotipo , Transducción de Señal , beta Catenina/metabolismo , Animales , Médula Ósea/metabolismo , Cartílago/citología , Células Cultivadas , Condrocitos/citología , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Medios de Cultivo/metabolismo , Citosol/metabolismo , Humanos , Hipertrofia/patología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Factor de Crecimiento Transformador beta3/farmacología , beta Catenina/genética
16.
Arthritis Rheum ; 63(9): 2711-20, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21547890

RESUMEN

OBJECTIVE: Human mesenchymal stem cells (hMSCs) represent one of the most promising stem cell therapies for traumatic injury and age-related degenerative diseases involving cartilage. However, few genetic factors regulating chondrogenesis of MSCs have been identified. One study showed that zinc-finger protein 145 (ZNF145), a transcription factor, was up-regulated during 3-lineage differentiation of hMSCs. The present study was undertaken to validate whether this novel transcription factor is useful for the repair and regeneration of cartilage. METHODS: Human MSCs were transfected with lentiviral short hairpin RNA (for small interfering RNA knockdown of ZNF145) and a lentiviral vector for overexpression of ZNF145, and the effects of ZNF145 on chondrogenesis were studied using quantitative polymerase chain reaction and immunostaining. Microarray and transient expression analyses were used to determine whether ZNF145 is a factor operating upstream of SOX9. Allogeneic transplantation of hMSCs into osteochondral defects in rats was performed to determine the effects of ZNF145 on repair of cartilage in vivo. RESULTS: Small interfering RNA-mediated gene silencing of ZNF145 slowed down chondrogenesis, whereas overexpression of ZNF145 enhanced chondrogenesis. Global gene expression profiling showed up-regulated gene expression in ZNF145-overexpressing MSCs, and transient overexpression of ZNF145 enhanced the expression of SOX9, suggesting that ZNF145 acts as a factor upstream of SOX9, the master regulator of chondrogenesis. Moreover, allogeneic transplantation of hMSCs into osteochondral defects of rat knees showed that ZNF145-overexpressing MSCs repaired cartilage defects better and earlier than empty control MSCs. CONCLUSION: These findings suggest that ZNF145 gene therapy may be a very useful strategy for improving the quality of cartilage regeneration and repair.


Asunto(s)
Cartílago/metabolismo , Condrogénesis/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Células Madre Mesenquimatosas/metabolismo , Factor de Transcripción SOX9/metabolismo , Animales , Linaje de la Célula , Condrocitos/metabolismo , Humanos , Masculino , Proteína de la Leucemia Promielocítica con Dedos de Zinc , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Regeneración/fisiología , Transducción de Señal/fisiología
17.
Stem Cells Dev ; 18(7): 1013-22, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19102659

RESUMEN

Mesenchymal stem cells (MSCs) represent a source of pluripotent cells that are already in various phases of clinical application. However, the use of MSCs in tissue engineering has been hampered largely due to their limitations, including low proliferation, finite life span, and gradual loss of their stem cell properties during ex vivo expansion. Nanog and Oct4 are key transcription factors essential to the pluripotent and self-renewing phenotypes of undifferentiated embryonic stem cells (ESCs). To determine whether Nanog and Oct4 improve human bone marrow-MSC quality, we therefore established stable Nanog and Oct4 overexpressing MSCs using a lentiviral system and showed that this promoted cell proliferation and enhanced colony formation of MSCs. In differentiating MSCs, Nanog, and Oct4, overexpression had converse effects on adipogenesis of MSCs and Nanog overexpression slowed down adipogenesis, whereas Oct4 overexpression improved adipogenesis. Nanog and Oct4 overexpression both improved chondrogenesis. Microarray data showed many differences in transcriptional targets in undifferentiated MSCs overexpressing Nanog and Oct4. These results provide insight into the improvement of the stemness of MSCs by genetic modification with stemness-related genes.


Asunto(s)
Expresión Génica , Proteínas de Homeodominio/biosíntesis , Células Madre Mesenquimatosas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Adipogénesis/genética , Proliferación Celular , Condrogénesis/genética , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Células Madre Mesenquimatosas/citología , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Ingeniería de Tejidos/métodos
18.
Stem Cells ; 25(3): 750-60, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17095706

RESUMEN

Mesenchymal stem cells derived from human bone marrow (hBMSCs) and human adipose tissue (hAMSCs) represent a useful source of progenitor cells for cell therapy and tissue engineering. However, it is not clear what the similarities and differences between them are. Like hBMSCs, hAMSCs can differentiate into osteogenic, adipogenic, and chondrogenic cells. Whether MSCs derived from different tissue sources represent fundamentally similar or different cell types is not clear. Given the possible different sources of MSCs for cell therapy, a comprehensive comparison of the different MSCs would be very useful. Here, we compared the transcriptome profile of hAMCS and hBMSCs during directed differentiation into bone, cartilage, and fat. Our data revealed considerable similarities between bone marrow-derived MSCs (BMSCs) and adipose tissue-derived MSCs (AMSCs). We uncovered an interesting bifurcation of pathways in both BMSCs and AMSCs, in which osteogenesis and adipogenesis appear to be linked in a differentiation branch separate from chondrogenesis. Our data suggest that although a set of common genes may be needed for early differentiation into all three lineages, a different set of signature genes is associated with maturation into fully differentiated cells. The recruitment of different late differentiation factors explains and supports our conclusion that BMSCs differentiate more efficiently into bone and cartilage, whereas AMSCs differentiate better into adipocytes. This study not only generated a rich database for continuing molecular characterization of various MSCs but also provided a rational basis for assessing qualities of MSCs from different sources for the purpose of cell-based therapy and tissue engineering.


Asunto(s)
Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Tejido Adiposo/citología , Tejido Adiposo/fisiología , Regulación de la Expresión Génica , Humanos , Immunoblotting , Osteogénesis , Reacción en Cadena de la Polimerasa , Interferencia de ARN , Regulación hacia Arriba
19.
Stem Cells ; 25(2): 425-36, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17053208

RESUMEN

Adult tissue-derived mesenchymal stem cells (MSCs) have demonstrated therapeutic efficacy in treating diseases or repairing damaged tissues through mechanisms thought to be mediated by either cell replacement or secretion of paracrine factors. Characterized, self-renewing human ESCs could potentially be an invariable source of consistently uniform MSCs for therapeutic applications. Here we describe a clinically relevant and reproducible manner of generating identical batches of hESC-derived MSC (hESC-MSC) cultures that circumvents exposure to virus, mouse cells, or serum. Trypsinization and propagation of HuES9 or H1 hESCs in feeder- and serum-free selection media generated three polyclonal, karyotypically stable, and phenotypically MSC-like cultures that do not express pluripotency-associated markers but displayed MSC-like surface antigens and gene expression profile. They differentiate into adipocytes, osteocytes, and chondrocytes in vitro. Gene expression and fluorescence-activated cell sorter analysis identified CD105 and CD24 as highly expressed antigens on hESC-MSCs and hESCs, respectively. CD105+, CD24- monoclonal isolates have a typical MSC gene expression profiles and were identical to each other with a highly correlated gene expression profile (r(2) > .90). We have developed a protocol to reproducibly generate clinically compliant and identical hESC-MSC cultures.


Asunto(s)
Antígenos CD/inmunología , Antígeno CD24/inmunología , Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Mesenquimatosas/citología , Receptores de Superficie Celular/inmunología , Adipogénesis , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígeno CD24/genética , Antígeno CD24/metabolismo , Separación Celular , Células Cultivadas , Condrogénesis , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/trasplante , Endoglina , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteogénesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Trasplante de Células Madre
20.
J Exp Zool ; 293(7): 719-25, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12410600

RESUMEN

Procedures to improve somatic cell nuclear transplantation in fish were evaluated. We reported effects of nonirradiated recipient eggs, inactivated recipient eggs, different combinations between recipient eggs and donor cells, duration of serum starvation, generation number, and passage number of donor cells on developmental rates of nuclear transplant (NT) embryos. Exposure to 25,000 R of gamma-rays inactivated recipient eggs. Single nucleus of cultured, synchronized somatic cell from gynogenetic bighead carp (Aristichthys nobilis) was transplanted into nonirradiated or genetically inactivated unfertilized egg of gibel carp (Carassius auratus gibelio). There was no significant difference in developmental rate between nonirradiated and inactivated recipient eggs (27.27% vs. 25.71%, respectively). Chromosome count showed that 70.59% of NT embryos contained 48 chromosomes. It showed that most NT embryos came from donor nuclei of bighead carp, which was supported by microsatellite analysis of NT embryos. But 23.53% of NT embryos contained more than 48 chromosomes. It was presumed that those superfluous chromosomes came from nonirradiated recipient eggs. Besides, 5.88% of NT embryos were chimeras. Eggs of blunt-snout bream (Megalobrama amblycephala) and gibel carp were better recipient eggs than those of loach (Misgurnus anguillicaudatus) (25% and 18.03% vs. 8.43%). Among different duration of serum starvation, developmental rate of NT embryos from somatic nuclei of three-day serum starvation was the highest, reaching 25.71% compared to 14.14% (control), 20% (five-day), and 21.95% (seven-day). Cultured donor cells of less passage facilitated reprogramming of NT embryos than those of more passage. Recloning might improve the developmental rate of NT embryos from the differentiated donor nuclei. Developmental rate of fourth generation was the highest (54.83%) and the lowest for first generation (14.14%) compared to second generation (38.96%) and third generation (53.01%).


Asunto(s)
Carpas/embriología , Clonación de Organismos/métodos , Embrión no Mamífero/citología , Desarrollo Embrionario , Técnicas de Transferencia Nuclear , Animales , Carpas/genética , Núcleo Celular/genética , Cromosomas/genética , Medio de Cultivo Libre de Suero , Femenino , Rayos gamma , Masculino , Repeticiones de Microsatélite/genética , Óvulo/citología , Óvulo/fisiología , Óvulo/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...