Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 112(2): 295-307, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18332228

RESUMEN

We compared the transcriptomes of marrow-derived mesenchymal stem cells (MSCs) with differentiated adipocytes, osteocytes, and chondrocytes derived from these MSCs. Using global gene-expression profiling arrays to detect RNA transcripts, we have identified markers that are specific for MSCs and their differentiated progeny. Further, we have also identified pathways that MSCs use to differentiate into adipogenic, chondrogenic, and osteogenic lineages. We identified activin-mediated transforming growth factor (TGF)-beta signaling, platelet-derived growth factor (PDGF) signaling and fibroblast growth factor (FGF) signaling as the key pathways involved in MSC differentiation. The differentiation of MSCs into these lineages is affected when these pathways are perturbed by inhibitors of cell surface receptor function. Since growth and differentiation are tightly linked processes, we also examined the importance of these 3 pathways in MSC growth. These 3 pathways were necessary and sufficient for MSC growth. Inhibiting any of these pathways slowed MSC growth, whereas a combination of TGF-beta, PDGF, and beta-FGF was sufficient to grow MSCs in a serum-free medium up to 5 passages. Thus, this study illustrates it is possible to predict signaling pathways active in cellular differentiation and growth using microarray data and experimentally verify these predictions.


Asunto(s)
Perfilación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/fisiología , Células Madre Mesenquimatosas/citología , Transducción de Señal , Adipocitos/citología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Condrocitos/citología , Factores de Crecimiento de Fibroblastos/fisiología , Humanos , Osteoblastos/citología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Factor de Crecimiento Transformador beta/fisiología
2.
Cytotherapy ; 11(7): 958-72, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19903107

RESUMEN

BACKGROUND AIMS: Human adipose stem cells (ASC) are an abundant, readily available population of multipotent progenitor cells that reside in adipose tissue. ASC have been shown to have therapeutic applicability in pre-clinical studies, but a standardized expansion method for clinical cell therapy has yet to be established. Isolated ASC are typically expanded in medium containing fetal bovine serum (FBS); however, sera and other culturing reagents of animal origin in clinical therapy pose numerous safety issues, including possible infections and severe immune reactions. METHODS: To identify optimal conditions for ex vivo expansion of ASC, the effects of seven serum-free (SF) and xeno-free (XF) media were investigated with both FBS and allogeneic human serum (alloHS; as a control media). Surface marker expression, proliferation, morphology and differentiation analyzes were utilized for investigating the effects of media on ASC. RESULTS: The proliferation and morphology analysis demonstrated significant differences between ASC cultured in SF/XF culture media compared with serum-containing culture media, with medium prototype StemPro MSC SFM XenoFree providing significantly higher proliferation rates than ASC cultured in media containing serum, while still maintaining the differentiation potential and surface marker expression profile characteristic of ASC. CONCLUSIONS: Looking forward, fully defined XF media formulations will provide the means for the development and approval of safer clinical cell therapy treatments. However, to fully recognize the capacity of these XF culture media, further pre-clinical safety and efficacy studies must be performed.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Medio de Cultivo Libre de Suero/metabolismo , Células Madre Multipotentes/metabolismo , Tejido Adiposo/citología , Antígenos de Diferenciación/metabolismo , Antígenos Heterófilos/inmunología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Persona de Mediana Edad , Células Madre Multipotentes/citología , Células Madre Multipotentes/inmunología
3.
Anal Biochem ; 393(1): 95-104, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19539597

RESUMEN

The activation of G-protein-coupled receptors (GPCRs) can result in the stimulation of numerous signaling networks that extend beyond canonical secondary messenger-dependent pathways. It is well-established that many of these diverse networks converge on the MAPK pathway, resulting in the activation of extracellular-signal regulated kinase 1/2 (ERK). Since the link between GPCRs and ERK can be modulated via both G-protein-dependent and -independent mechanisms, measurement of ERK phosphorylation may serve as an ideal surrogate for GPCR activation. We have combined BacMam-mediated gene delivery of the GFP-ERK2 with a time-resolved Foerster resonance energy transfer (TR-FRET) immunoassay for the measurement of intracellular phospho-ERK2 levels. Together these technologies enable a flexible platform for measuring GPCR and MAPK activation in the cell line of interest. This technology has been applied to the measurement of activation of the serotonin 5-hydroxytryptamine-1A (5-HT(1A)) receptor expressed in CHO-K1 cells. In addition to demonstrating the flexibility of this assay platform, we provide the first reported profile for 5-HT(1A) receptor-mediated ERK activation using a panel of known Parkinson's disease drugs. Our results demonstrate the value of using ERK activation as a downstream sensor for GPCR function, providing an attractive complement to upstream endpoints such as ligand occupancy and binding of GTPgammaS.


Asunto(s)
Técnicas Biosensibles/métodos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Fosfoproteínas/metabolismo , Receptor de Serotonina 5-HT1A/análisis , Receptor de Serotonina 5-HT1A/metabolismo , Animales , Línea Celular , Cricetinae , Humanos , Receptor de Serotonina 5-HT1A/genética
4.
Exp Hematol ; 36(10): 1354-1369, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18657893

RESUMEN

OBJECTIVE: Human multipotent mesenchymal stromal cells (MSC) have the potential to differentiate into multiple cell types, although little is known about factors that control their fate. Differentiation-specific microRNAs may play a key role in stem cell self-renewal and differentiation. We propose that specific intracellular signaling pathways modulate gene expression during differentiation by regulating microRNA expression. MATERIALS AND METHODS: Illumina mRNA and NCode microRNA expression analyses were performed on MSC and their differentiated progeny. A combination of bioinformatic prediction and pathway inhibition was used to identify microRNAs associated with platelet-derived growth factor (PDGF) signaling. RESULTS: The pattern of microRNA expression in MSC is distinct from that in pluripotent stem cells, such as human embryonic stem cells. Specific populations of microRNAs are regulated in MSC during differentiation targeted toward specific cell types. Complementary mRNA expression analysis increases the pool of markers characteristic of MSC or differentiated progeny. To identify microRNA expression patterns affected by signaling pathways, we examined the PDGF pathway found to be regulated during osteogenesis by microarray studies. A set of microRNAs bioinformatically predicted to respond to PDGF signaling was experimentally confirmed by direct PDGF inhibition. CONCLUSION: Our results demonstrate that a subset of microRNAs regulated during osteogenic differentiation of MSCs is responsive to perturbation of the PDGF pathway. This approach not only identifies characteristic classes of differentiation-specific mRNAs and microRNAs, but begins to link regulated molecules with specific cellular pathways.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/citología , MicroARNs/genética , Osteogénesis/fisiología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Células Madre Pluripotentes/citología , Adipocitos/citología , Adolescente , Adulto , Población Negra , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Condrocitos/citología , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
5.
Sci Transl Med ; 11(475)2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30651323

RESUMEN

Considerable progress has been made in testing stem cell-derived retinal pigment epithelium (RPE) as a potential therapy for age-related macular degeneration (AMD). However, the recent reports of oncogenic mutations in induced pluripotent stem cells (iPSCs) underlie the need for robust manufacturing and functional validation of clinical-grade iPSC-derived RPE before transplantation. Here, we developed oncogenic mutation-free clinical-grade iPSCs from three AMD patients and differentiated them into clinical-grade iPSC-RPE patches on biodegradable scaffolds. Functional validation of clinical-grade iPSC-RPE patches revealed specific features that distinguished transplantable from nontransplantable patches. Compared to RPE cells in suspension, our biodegradable scaffold approach improved integration and functionality of RPE patches in rats and in a porcine laser-induced RPE injury model that mimics AMD-like eye conditions. Our results suggest that the in vitro and in vivo preclinical functional validation of iPSC-RPE patches developed here might ultimately be useful for evaluation and optimization of autologous iPSC-based therapies.


Asunto(s)
Degeneración Retiniana/terapia , Epitelio Pigmentado de la Retina/citología , Células Madre/citología , Animales , Modelos Animales de Enfermedad , Degeneración Macular/patología , Degeneración Macular/terapia , Ratas , Degeneración Retiniana/patología , Porcinos
6.
Diabetes ; 56(1): 3-7, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17110468

RESUMEN

As recent studies suggest that newly formed pancreatic beta-cells are a result of self-duplication rather than stem cell differentiation, in vitro expansion of beta-cells presents a potential mechanism by which to increase available donor tissue for cell-based diabetes therapies. Although most studies have found that beta-cells are resilient to substantial in vitro expansion, recent studies have suggested that it is possible to expand these cells through a process referred to as epithelial-mesenchymal transition (EMT). To further substantiate such an expansion mechanism, we used recombination-based genetic lineage tracing to determine the origin of proliferating fibroblast-like cells from cultured pancreatic islets in vitro. We demonstrate, using two culture methods, that EMT does not underlie the appearance of fibroblast-like cells in mouse islet cultures but that fibroblast-like cells appear to represent mesenchymal stem cell (MSC)-like cells akin to MSCs isolated from bone marrow.


Asunto(s)
Células Epiteliales/fisiología , Fibroblastos/citología , Fibroblastos/fisiología , Proteínas de Homeodominio/fisiología , Insulina/fisiología , Islotes Pancreáticos/citología , Mesodermo/fisiología , Transactivadores/fisiología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , División Celular , ADN Complementario/genética , Citometría de Flujo , Receptores de Hialuranos/análisis , Islotes Pancreáticos/metabolismo , Antígenos Comunes de Leucocito/análisis , Mesodermo/citología , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , ARN/genética , ARN/aislamiento & purificación
7.
Curr Opin Chem Biol ; 11(4): 367-72, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17692558

RESUMEN

Human embryonic stem cells, because of their unique combination of long-term self-renewal properties and pluripotency, are providing new avenues of investigation of stem cell biology and human development and show promise in providing a new source of human cells for transplantation therapies and pharmaceutical testing. Current methods of propagating these cells using combinations of mouse fibroblast feeder cultures and bovine serum components are inexpensive and, in general, useful. However, the systematic investigation of the regulation of self-renewal and the production of safer sources of cells for transplantation depends on the elimination of animal products and the use of defined culture conditions. Both goals are served by the development of serum-free culture methods for human embryonic stem cells.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Embrionarias/citología , Animales , Supervivencia Celular , Medio de Cultivo Libre de Suero , Humanos
8.
Liver Transpl ; 14(3): 333-45, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18306374

RESUMEN

Widespread use of liver transplantation in the treatment of hepatic diseases is restricted by the limited availability of donated organs. One potential solution to this problem would be isolation and propagation of liver progenitor cells or stem cells. Here, we report on the isolation of a novel progenitor cell population from unmanipulated (that is, no prior exposure to chemicals and no injury) adult rat liver. Rat liver cells were cultured following a protocol developed in our laboratory to generate a unique progenitor cell population called liver-derived progenitor cells (LDPCs). LDPCs were analyzed by fluorescence-activated cell sorting, real-time polymerase chain reaction (RT-PCR), immunostaining and microarray gene expression. LDPCs were also differentiated into hepatocytes and biliary epithelium in vitro and examined for mature hepatic markers and urea and albumin production. These analyses showed that, LDPCs expressed stem cell markers such as cluster domain (CD)45, CD34, c-kit, and Thy 1, similar to hematopoietic stem cells, as well as endodermal/hepatic markers such as hepatocyte nuclear factor (HNF)3beta, hematopoietically-expressed homeobox gene-1, c-met, and transthyretin. LDPCs were negative for OV-6, cytokeratins (CKs), albumin, and HNF1alpha. The microarray gene expression profile demonstrated that they showed some similarities to known liver progenitor/stem cells such as oval cells. In addition, LDPCs differentiated into functional hepatocytes in vitro as shown by albumin expression and urea production. In conclusion, LDPCs are a population of unique liver progenitors that can be generated from unmanipulated adult liver, which makes them potentially useful for clinical applications, especially for cell transplantation in the treatment of liver diseases.


Asunto(s)
Separación Celular/métodos , Hígado/citología , Células Madre/citología , Células Madre/metabolismo , Albúminas/metabolismo , Animales , Conductos Biliares/citología , Conductos Biliares/metabolismo , Diferenciación Celular , Células Cultivadas , Citometría de Flujo , Hepatocitos/citología , Hepatocitos/metabolismo , Cariotipificación , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/metabolismo , Hígado/metabolismo , Análisis por Micromatrices , Prealbúmina/genética , Prealbúmina/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Urea/metabolismo
9.
Pathol Oncol Res ; 13(4): 302-10, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18158565

RESUMEN

The Notch signaling pathway is a multifunctional, evolutionarily conserved pathway, which plays an important role in development as well as stem cell biology. Multipotent adult progenitor cells (MAPCs) represent a unique stem cell population, which is capable of differentiating into cell types of the ectodermal, mesodermal and endodermal lineages in vitro, and contribute to most somatic cell types in vivo. Our aim was to characterize the gene expression of Notch signaling elements in rodent MAPCs. We show that transcripts for Notch-receptors, ligands, regulatory molecules of the pathway and the Hairy/Enhancer of Split-1 (HES-1) target gene are present in mouse and rat low-Oct4 MAPCs. We found that mouse Notch3 and rat Notch1 transcripts increased when cells were cultured at high density for 48 to 96 hours. HES-1 and HES-related transcription factor-1 (HERP-1), transcriptional targets of Notch-signaling, were both elicited by immobilized Delta1 ligand. In addition, mRNA for Notch1 and Notch3 was also induced by Notch-signaling, suggesting the presence of regulatory feedback loops. Slight differences between mouse and rat derived MAPCs suggest that the exact function, transcriptional regulation and the fine-tuning of the signal may be species specific. Taken together, we characterized the gene expression profile of the Notch pathway in rodent low-Oct4-MAPCs, and showed that the pathway is functional and can be modulated. Our results provide an additional tool and a further basis for a better understanding of stem cell biology.


Asunto(s)
Células Madre Multipotentes/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Transcripción Genética , Animales , Células Cultivadas , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/metabolismo , Ratones , Células Madre Multipotentes/citología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Ratas , Receptores Notch/genética
10.
Tissue Eng Part C Methods ; 18(8): 593-602, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22364177

RESUMEN

Development and implementation of therapeutic protocols based on stem cells or tissue-engineered products relies on methods that enable the production of substantial numbers of cells while complying with stringent quality and safety demands. In the current study, we aimed to assess the benefits of maintaining cultures of adipose-derived stem cells (ASCs) in a defined culture system devoid of xenogeneic components (xeno-free) and hypoxia over a 49-day growth period. Our data provide evidence that conditions involving StemPro mesenchymal stem cells serum-free medium (SFM) Xeno-Free and hypoxia (5% oxygen concentration) in the culture atmosphere provide a superior proliferation rate compared to a standard growth environment comprised of alpha-modified Eagle medium (A-MEM) supplemented with fetal calf serum (FCS) and ambient air (20% oxygen concentration) or that of A-MEM supplemented with FCS and hypoxia. Furthermore, a flow cytometric analysis and in vitro differentiation assays confirmed the immunophenotype stability and maintained multipotency of ASCs when expanded under xeno-free conditions and hypoxia. In conclusion, our data demonstrate that growth conditions utilizing a xeno-free and hypoxic environment not only provide an improved environment for the expansion of ASCs, but also set the stage as a culture system with the potential broad spectrum utility for regenerative medicine and tissue engineering applications.


Asunto(s)
Tejido Adiposo/citología , Técnicas de Cultivo de Célula/métodos , Células Madre/citología , Diferenciación Celular , Proliferación Celular , Medios de Cultivo/farmacología , Medio de Cultivo Libre de Suero/farmacología , Citometría de Flujo/métodos , Humanos , Hipoxia , Inmunofenotipificación/métodos , Oxígeno/química , Medicina Regenerativa/métodos , Ingeniería de Tejidos/métodos
11.
Stem Cells Transl Med ; 1(10): 750-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23197667

RESUMEN

Human multipotent mesenchymal stem cell (MSC) therapies are currently being tested in clinical trials for Crohn's disease, multiple sclerosis, graft-versus-host disease, type 1 diabetes, bone fractures, cartilage damage, and cardiac diseases. Despite remarkable progress in clinical trials, most applications still use traditional culture media containing fetal bovine serum or serum-free media that contain serum albumin, insulin, and transferrin. The ill-defined and variable nature of traditional culture media remains a challenge and has created a need for better defined xeno-free culture media to meet the regulatory and long-term safety requirements for cell-based therapies. We developed and tested a serum-free and xeno-free culture medium (SFM-XF) using human bone marrow- and adipose-derived MSCs by investigating primary cell isolation, multiple passage expansion, mesoderm differentiation, cellular phenotype, and gene expression analysis, which are critical for complying with translation to cell therapy. Human MSCs expanded in SFM-XF showed continual propagation, with an expected phenotype and differentiation potential to adipogenic, chondrogenic, and osteogenic lineages similar to that of MSCs expanded in traditional serum-containing culture medium (SCM). To monitor global gene expression, the transcriptomes of bone marrow-derived MSCs expanded in SFM-XF and SCM were compared, revealing relatively similar expression profiles. In addition, the SFM-XF supported the isolation and propagation of human MSCs from primary human marrow aspirates, ensuring that these methods and reagents are compatible for translation to therapy. The SFM-XF culture system allows better expansion and multipotentiality of MSCs and serves as a preferred alternative to serum-containing media for the production of large scale, functionally competent MSCs for future clinical applications.


Asunto(s)
Técnicas de Cultivo de Célula/normas , Diferenciación Celular , Medio de Cultivo Libre de Suero/química , Células Madre Mesenquimatosas/citología , Células Madre Multipotentes/citología , Tejido Adiposo/química , Tejido Adiposo/citología , Médula Ósea/química , Técnicas de Cultivo de Célula/métodos , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Condrogénesis , Medio de Cultivo Libre de Suero/normas , Citometría de Flujo , Inestabilidad Genómica , Humanos , Inmunofenotipificación , Cariotipificación , Células Madre Mesenquimatosas/química , Células Madre Multipotentes/química , Fenotipo , Transcriptoma
12.
Toxicol Sci ; 126(2): 426-35, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22223483

RESUMEN

Human induced pluripotent stem cells (hiPSC) hold great promise for providing various differentiated cell models for in vitro toxigenicity testing. For Clostridium botulinum neurotoxin (BoNT) detection and mechanistic studies, several cell models currently exist, but none examine toxin function with species-specific relevance while exhibiting high sensitivity. The most sensitive cell models to date are mouse or rat primary cells and neurons derived from mouse embryonic stem cells, both of which require significant technical expertise for culture preparation. This study describes for the first time the use of hiPSC-derived neurons for BoNT detection. The neurons used in this study were differentiated and cryopreserved by Cellular Dynamics International (Madison, WI) and consist of an almost pure pan-neuronal population of predominantly gamma aminoisobutyric acidergic and glutamatergic neurons. Western blot and quantitative PCR data show that these neurons express all the necessary receptors and substrates for BoNT intoxication. BoNT/A intoxication studies demonstrate that the hiPSC-derived neurons reproducibly and quantitatively detect biologically active BoNT/A with high sensitivity (EC(50) ∼0.3 U). Additionally, the quantitative detection of BoNT serotypes B, C, E, and BoNT/A complex was demonstrated, and BoNT/A specificity was confirmed through antibody protection studies. A direct comparison of BoNT detection using primary rat spinal cord cells and hiPSC-derived neurons showed equal or increased sensitivity, a steeper dose-response curve and a more complete SNARE protein target cleavage for hiPSC-derived neurons. In summary, these data suggest that neurons derived from hiPSCs provide an ideal and highly sensitive platform for BoNT potency determination, neutralizing antibody detection and for mechanistic studies.


Asunto(s)
Toxinas Botulínicas Tipo A/análisis , Neuronas/citología , Células Madre Pluripotentes/citología , Animales , Células Cultivadas , Humanos , Ratones , Ratas
13.
Methods Mol Biol ; 698: 3-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21431506

RESUMEN

Research on mesenchymal stem cells (MSC) is progressing with increasing popularity. Currently there are a significant number of clinical trials exploring the use of MSCs for the treatment of various diseases including graft-versus-host disease, Crohn's disease, myocardial infarction, stroke, bone defects, diabetes, and wound repair (www.-clinicaltrials.gov). At the same time, there are questions associated with MSCs in terms of their isolation, culture expansion, phenotype, multipotential differentiation, and transplantation efficiency. This chapter outlines the current status of the field and emphasizes the need for clearly defined protocols to better define the function and use of MSCs in cell therapy.


Asunto(s)
Técnicas Citológicas/métodos , Células Madre Mesenquimatosas/citología , Animales , Diferenciación Celular , Separación Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos
14.
Methods Mol Biol ; 698: 459-84, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21431538

RESUMEN

Human mesenchymal stem cells (hMSCs) are multipotent stem cells that can differentiate into a variety of cell types in vitro including osteoblasts, adipocytes, and chondrocytes. Here we apply a metabolic labeling approach to characterize changes in cellular glycoprotein expression during hMSC differentiation and to identify glycoprotein markers unique to differentiated cell types. The two-step labeling method involves the metabolic incorporation of unnatural azido-modified sugars into protein glycans and subsequent ligation with fluorescent azide-reactive detection probes utilizing the copper (I)-catalyzed cycloaddition reaction between azides and alkynes, or "click" chemistry. Metabolic labeling of cell surface O-linked or sialic acid-containing glycoproteins, or intracellular O-GlcNAc-modified proteins was accomplished by feeding cells the tetraacetylated azide-modified sugar precursors, GalNAz, ManNAz, or GlcNAz, respectively, for 48-72 h prior to harvesting the cells. The cells were then lysed, and protein extracts were reacted with a fluorescent alkyne detection probe. Labeled glycoproteins were analyzed by 1D and 2D gel electrophoresis and detected by fluorescence imaging. Our results demonstrate highly sensitive labeling of O-linked, sialic acid-containing, and O-GlcNAc modified proteins in all cell types without affecting cell growth or morphology. Selective labeling of sialic acid-containing glycoproteins by ManNAz was validated by loss of labeling following digestion with sialidase A. Significant changes in cellular glycoprotein profiles were seen upon differentiation into different cell types, and several putative glycoprotein markers were identified by MALDI peptide fingerprinting. One of these identified proteins, Galectin 1, is validated and shown for the first time to be posttranslationally modified by O-glycosylation, most likely by O-linked N-acetylglucosamine (O-GlcNAc).


Asunto(s)
Diferenciación Celular , Química Clic , Glicoproteínas/química , Glicoproteínas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Coloración y Etiquetado/métodos , Adipocitos/citología , Adipocitos/metabolismo , Azul Alcián/metabolismo , Antraquinonas/metabolismo , Compuestos Azo/metabolismo , Biomarcadores/química , Biomarcadores/metabolismo , Western Blotting , Técnicas de Cultivo de Célula , Condrocitos/citología , Condrocitos/metabolismo , Electroforesis en Gel Bidimensional , Glicósido Hidrolasas/metabolismo , Humanos , Focalización Isoeléctrica , Espectrometría de Masas , Osteoblastos/citología , Osteoblastos/metabolismo
15.
Tissue Eng Part C Methods ; 17(12): 1201-10, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21895491

RESUMEN

The immunomodulatory properties of mesenchymal stem cells (MSCs) make them attractive therapeutic agents for a wide range of diseases. However, the highly demanding cell doses used in MSC clinical trials (up to millions of cells/kg patient) currently require labor intensive methods and incur high reagent costs. Moreover, the use of xenogenic (xeno) serum-containing media represents a risk of contamination and raises safety concerns. Bioreactor systems in combination with novel xeno-free medium formulations represent a viable alternative to reproducibly achieve a safe and reliable MSC doses relevant for cell therapy. The main goal of the present study was to develop a complete xeno-free microcarrier-based culture system for the efficient expansion of human MSC from two different sources, human bone marrow (BM), and adipose tissue. After 14 days of culture in spinner flasks, BM MSC reached a maximum cell density of (2.0±0.2)×105 cells·mL⁻¹ (18±1-fold increase), whereas adipose tissue-derived stem cells expanded to (1.4±0.5)×105 cells·mL⁻¹ (14±7-fold increase). After the expansion, MSC expressed the characteristic markers CD73, CD90, and CD105, whereas negative for CD80 and human leukocyte antigen (HLA)-DR. Expanded cells maintained the ability to differentiate robustly into osteoblast, adipocyte, and chondroblast lineages upon directed differentiation. These results demonstrated the feasibility of expanding human MSC in a scalable microcarrier-based stirred culture system under xeno-free conditions and represent an important step forward for the implementation of a Good Manufacturing Practices-compliant large-scale production system of MSC for cellular therapy.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Mesenquimatosas/citología , Microesferas , Tejido Adiposo/citología , Adulto , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medios de Cultivo/farmacología , Humanos , Inmunofenotipificación , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Plásticos
16.
Stem Cell Res Ther ; 1(1): 8, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20504289

RESUMEN

INTRODUCTION: Human multipotent mesenchymal stem cell (MSC) therapies are being tested clinically for a variety of disorders, including Crohn's disease, multiple sclerosis, graft-versus-host disease, type 1 diabetes, bone fractures, and cartilage defects. However, despite the remarkable clinical advancements in this field, most applications still use traditional culture media containing fetal bovine serum. The ill-defined and highly variable nature of traditional culture media remains a challenge, hampering both the basic and clinical human MSC research fields. To date, no reliable serum-free medium for human MSCs has been available. METHODS: In this study, we developed and tested a serum-free growth medium on human bone marrow-derived MSCs through the investigation of multiple parameters including primary cell isolation, multipassage expansion, mesoderm differentiation, cellular phenotype, and gene-expression analysis. RESULTS: Similar to that achieved with traditional culture medium, human MSCs expanded in serum-free medium supplemented with recombinant human platelet-derived growth factor-BB (PDGF-BB), basic fibroblast growth factor (bFGF), and transforming growth factor (TGF)-beta1 showed extensive propagation with retained phenotypic, differentiation, and colony-forming unit potential. To monitor global gene expression, the transcriptomes of bone marrow-derived MSCs expanded under serum-free and serum-containing conditions were compared, revealing similar expression profiles. In addition, the described serum-free culture medium supported the isolation of human MSCs from primary human marrow aspirate with continual propagation. CONCLUSIONS: Although the described serum-free MSC culture medium is not free of xenogeneic components, this medium provides a substitute for serum-containing medium for research applications, setting the stage for future clinical applications.


Asunto(s)
Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medios de Cultivo/química , Células Madre Mesenquimatosas/metabolismo , Becaplermina , Células Cultivadas , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/citología , Proteínas Proto-Oncogénicas c-sis/metabolismo , Suero , Factor de Crecimiento Transformador beta1/metabolismo
17.
Genome Biol ; 8(8): R163, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17683608

RESUMEN

BACKGROUND: Recently, several populations of postnatal stem cells, such as multipotent adult progenitor cells (MAPCs), have been described that have broader differentiation ability than classical adult stem cells. Here we compare the transcriptome of pluripotent embryonic stem cells (ESCs), MAPCs, and lineage-restricted mesenchymal stem cells (MSCs) to determine their relationship. RESULTS: Applying principal component analysis, non-negative matrix factorization and k-means clustering algorithms to the gene-expression data, we identified a unique gene-expression profile for MAPCs. Apart from the ESC-specific transcription factor Oct4 and other ESC transcripts, some of them associated with maintaining ESC pluripotency, MAPCs also express transcripts characteristic of early endoderm and mesoderm. MAPCs do not, however, express Nanog or Sox2, two other key transcription factors involved in maintaining ESC properties. This unique molecular signature was seen irrespective of the microarray platform used and was very similar for both mouse and rat MAPCs. As MSC-like cells isolated under MAPC conditions are virtually identical to MSCs, and MSCs cultured in MAPC conditions do not upregulate MAPC-expressed transcripts, the MAPC signature is cell-type specific and not merely the result of differing culture conditions. CONCLUSION: Multivariate analysis techniques clustered stem cells on the basis of their expressed gene profile, and the genes determining this clustering reflected the stem cells' differentiation potential in vitro. This comparative transcriptome analysis should significantly aid the isolation and culture of MAPCs and MAPC-like cells, and form the basis for studies to gain insights into genes that confer on these cells their greater developmental potency.


Asunto(s)
Células Madre Adultas/metabolismo , Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Células Madre Multipotentes/metabolismo , Transcripción Genética , Células Madre Adultas/citología , Animales , Medios de Cultivo/farmacología , Células Madre Embrionarias/citología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Células Madre Multipotentes/citología , Células Madre Multipotentes/efectos de los fármacos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA