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
1.
Mol Cell Biochem ; 476(7): 2813-2821, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33733429

RESUMEN

Environmental changes can stress and alter biology at the molecular and cellular level. For example, metal-protein interaction is a classic physic and biological property of nature, which is fundamentally influenced by acidity. Here, we report a unique cellular reprogramming phenomenon in that a brief strong acid treatment induced the expression of pluripotent stem cell (PSC) markers. We used strong acid to briefly challenge mix-cultured gastric cells, and then subcultured survived cells in a normal cell culture medium. We found that survival acid-treated cells expressed PSC markers detected by commonly used pluripotent antibodies such as SSEA-4 and Oct4. In addition, we observed that the survived cells from the acid challenge grew faster during the second and third weeks of subculture and had a relative short doubling time (DT) than the controls. PSC marker-labeled 'older' cells also presented immature cell-like morphology with some having marker Oct4 in the nucleus. Finally, the expression of the markers appeared to be sensitive to metal ion chelation. Removal of the metals during a brief acid treatment reduced pluripotent marker-positive cells, suggesting the dissociation of metals from metal-binding proteins may be a factor involved in the induction of stem cell markers. Our findings reveal that somatic cells appear to possess a plasticity feature to express pluripotent marker proteins or to select cell subpopulations that express pluripotent marker proteins when cells are transiently exposed to strong acid. It opens new directions for understanding conserved regulatory mechanisms involved in cellular survival under stressful stimulation.


Asunto(s)
Mucosa Gástrica/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ácido Clorhídrico/farmacología , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Antígenos Embrionarios Específico de Estadio/biosíntesis , Animales , Células Cultivadas , Células HeLa , Humanos , Ratones
2.
Sci Rep ; 6: 37004, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27845370

RESUMEN

Oral submucous fibrosis (OSF), regarded as a precancerous condition, is characterized by juxta-epithelial inflammatory reaction followed by fibro-elastic change in the lamina properia and epithelial atrophy. The pathologic mechanisms of OSF still need to be further clarified. In the study, we investigated the functional expression of SSEA-4, which is a well-known stemness marker, in myofibroblast activity and the clinical significance in OSF tissues. The expression of SSEA-4 in OSF was evaluated by immunohistochemical staining. Functional analysis of SSEA-4 on myofibroblast activity of OSF was achieved by lentiviral silencing ST3GAL2. Immunohisitochemistry demonstrated that SSEA-4 expression was significantly higher expression in areca quid chewing-associated OSF tissues than those of normal oral mucosa tissues. From flow cytometry analysis, arecoline dose-dependently activated SSEA-4 expression in primary human normal buccal mucosal fibroblasts (BMFs). Sorted SSEA-4-positive cells from fibrotic BMFs (fBMFs) have higher colony-forming unit, collagen gel contraction, and α-smooth muscle actin (α-SMA) expression than SSEA-4-negative subset. Knockdown of ST3GAL2 in fBMFs suppressed SSEA-4 expression, collagen contraction, migration, invasiveness, and wound healing capability. Consistently, silencing ST3GAL2 was found to repress arecoline-induced myofibroblast activity in BMFs. The study highlights SSEA-4 as a critical marker for therapeutic intervention to mediate myofibroblast transdifferentiation in areca quid chewing-associated OSF.


Asunto(s)
Biomarcadores de Tumor , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Boca , Miofibroblastos , Lesiones Precancerosas , Antígenos Embrionarios Específico de Estadio , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Femenino , Humanos , Masculino , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Antígenos Embrionarios Específico de Estadio/biosíntesis , Antígenos Embrionarios Específico de Estadio/genética
3.
Cell Reprogram ; 18(2): 67-77, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27055628

RESUMEN

A new type of mesenchymal stem cells (MSCs) that expresses stage-specific embryonic antigen 3 (SSEA-3) and the mesenchymal cell marker CD105 are known as multilineage-differentiating stress-enduring (Muse) cells. Studies have shown that stem cells in suspension cultures are more likely to generate embryoid body-like stem cell spheres and maintain an undifferentiated phenotype and pluripotency. We separated Muse cells derived from human dermal fibroblasts by long-term trypsin incubation (LTT) through suspension cultures in methylcellulose. The Muse cells obtained expressed several pluripotency markers, including Nanog, Oct4, Sox2, and SSEA-3, and could differentiate in vitro into cells of the three germ layers, such as hepatocytes (endodermal), neural cells (ectodermal) and adipocytes, and osteocytes (mesodermal cells). These cells showed a low level of DNA methylation and a high nucleo-cytoplasmic ratio. Our study provides an innovative and exciting platform for exploring the potential cell-based therapy of various human diseases using Muse cells as well as their great possibility for regenerative medicine.


Asunto(s)
Reprogramación Celular , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Antígenos de Diferenciación/biosíntesis , Antígenos de Carbohidratos Asociados a Tumores/biosíntesis , Endoglina/biosíntesis , Fibroblastos/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Mesenquimatosas/citología , Antígenos Embrionarios Específico de Estadio/biosíntesis
4.
In Vitro Cell Dev Biol Anim ; 52(2): 218-27, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26487430

RESUMEN

Gingival stem cells (GSCs) are a novel source of mesenchymal stem cells (MSCs) that are easily accessed from the oral cavity. GSCs were considered valuable autograft MSCs with particular characteristics. However, the limitation in the number of available GSCs remains an obstacle. Therefore, this study aimed to stimulate GSC proliferation by ascorbic acid (AA) and determined the effects of AA on GSC pluripotent potential-related gene expression. GSCs were isolated from gum tissue by explant culture and continuously subcultured before analysis of stemness and effects of AA on pluripotent-related gene expression. GSCs cultured with various concentrations of AA showed increased proliferation in a dose-dependent manner. AA-treated GSCs showed significantly higher expression of SSEA-3, Sox-2, Oct-3/4, Nanog, and TRA-1-60 compared with control cells. More importantly, GSCs also maintained their stemness with MSC phenotypes and failed to cause tumors in nude athymic mice. Our results show that AA is a suitable factor to stimulate GSC proliferation.


Asunto(s)
Ácido Ascórbico/administración & dosificación , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Encía/citología , Células Madre Mesenquimatosas/citología , Animales , Antígenos de Superficie/biosíntesis , Antígenos de Carbohidratos Asociados a Tumores/biosíntesis , Biomarcadores/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Encía/efectos de los fármacos , Encía/crecimiento & desarrollo , Proteínas de Homeodominio/biosíntesis , Humanos , Ratones , Boca/citología , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Proteoglicanos/biosíntesis , Factores de Transcripción SOXB1/biosíntesis , Antígenos Embrionarios Específico de Estadio/biosíntesis
5.
Stem Cells Transl Med ; 4(2): 146-55, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25561682

RESUMEN

Stage-specific embryonic antigen-3 (SSEA-3)-positive multipotent mesenchymal cells (multilineage differentiating stress-enduring [Muse] cells) were isolated from cultured human adipose tissue-derived stem/stromal cells (hASCs) and characterized, and their therapeutic potential for treating diabetic skin ulcers was evaluated. Cultured hASCs were separated using magnetic-activated cell sorting into positive and negative fractions, a SSEA-3+ cell-enriched fraction (Muse-rich) and the remaining fraction (Muse-poor). Muse-rich hASCs showed upregulated and downregulated pluripotency and cell proliferation genes, respectively, compared with Muse-poor hASCs. These cells also released higher amounts of certain growth factors, particularly under hypoxic conditions, compared with Muse-poor cells. Skin ulcers were generated in severe combined immunodeficiency (SCID) mice with type 1 diabetes, which showed delayed wound healing compared with nondiabetic SCID mice. Treatment with Muse-rich cells significantly accelerated wound healing compared with treatment with Muse-poor cells. Transplanted cells were integrated into the regenerated dermis as vascular endothelial cells and other cells. However, they were not detected in the surrounding intact regions. Thus, the selected population of ASCs has greater therapeutic effects to accelerate impaired wound healing associated with type 1 diabetes. These cells can be achieved in large amounts with minimal morbidity and could be a practical tool for a variety of stem cell-depleted or ischemic conditions of various organs and tissues.


Asunto(s)
Tejido Adiposo/metabolismo , Antígenos de Carbohidratos Asociados a Tumores/biosíntesis , Complicaciones de la Diabetes/terapia , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Células Madre Multipotentes/metabolismo , Úlcera Cutánea/terapia , Antígenos Embrionarios Específico de Estadio/biosíntesis , Tejido Adiposo/patología , Adolescente , Adulto , Animales , Complicaciones de la Diabetes/metabolismo , Complicaciones de la Diabetes/patología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Femenino , Xenoinjertos , Humanos , Ratones SCID , Úlcera Cutánea/metabolismo , Úlcera Cutánea/patología , Cicatrización de Heridas
6.
Immunity ; 41(5): 753-61, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25464854

RESUMEN

Medullary thymic epithelial cells (mTECs) are crucial for central T cell self-tolerance. Although progenitors of mTECs have been demonstrated in thymic organogenesis, the mechanism for postnatal mTEC maintenance remains elusive. We demonstrate that implantation of embryonic TECs expressing claudin-3 and claudin-4 (Cld3,4) in a medulla-defective thymic microenvironment restores medulla formation and suppresses multiorgan autoimmunity throughout life. A minor SSEA-1(+) fraction within the embryonic Cld3,4(hi) TECs contained self-renewable clonogenic TECs, capable of preferentially generating mature mTECs in vivo. Adult SSEA-1(+)Cld3,4(hi) TECs retained mTEC reconstitution potential, although the activity decreased. The clonogenicity of TECs also declined rapidly after birth in wild-type mice, whereas it persisted in Rag2(?/?) adult mice with defective thymopoiesis. The results suggest that unipotent mTEC-restricted stem cells that develop in the embryo have the capacity to functionally reconstitute the thymic medulla long-term, thus ensuring lifelong central T cell self-tolerance.


Asunto(s)
Organogénesis/inmunología , Autotolerancia/inmunología , Células Madre/inmunología , Linfocitos T/inmunología , Timo/citología , Animales , Antígenos de Carbohidratos Asociados a Tumores/biosíntesis , Diferenciación Celular/inmunología , Células Cultivadas , Claudina-3/biosíntesis , Claudina-4/biosíntesis , Proteínas de Unión al ADN/genética , Células Epiteliales/citología , Células Epiteliales/inmunología , Antígeno Lewis X/biosíntesis , Ratones , Ratones Noqueados , Antígenos Embrionarios Específico de Estadio/biosíntesis , Células Madre/citología , Timo/inmunología
7.
J Vis Exp ; (68)2012 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-23149977

RESUMEN

Through the ectopic expression of four transcription factors, Oct4, Klf4, Sox2 and cMyc, human somatic cells can be converted to a pluripotent state, generating so-called induced pluripotent stem cells (iPSCs)(1-4). Patient-specific iPSCs lack the ethical concerns that surround embryonic stem cells (ESCs) and would bypass possible immune rejection. Thus, iPSCs have attracted considerable attention for disease modeling studies, the screening of pharmacological compounds, and regenerative therapies(5). We have shown the generation of transgene-free human iPSCs from patients with different lung diseases using a single excisable polycistronic lentiviral Stem Cell Cassette (STEMCCA) encoding the Yamanaka factors(6). These iPSC lines were generated from skin fibroblasts, the most common cell type used for reprogramming. Normally, obtaining fibroblasts requires a skin punch biopsy followed by expansion of the cells in culture for a few passages. Importantly, a number of groups have reported the reprogramming of human peripheral blood cells into iPSCs(7-9). In one study, a Tet inducible version of the STEMCCA vector was employed(9), which required the blood cells to be simultaneously infected with a constitutively active lentivirus encoding the reverse tetracycline transactivator. In contrast to fibroblasts, peripheral blood cells can be collected via minimally invasive procedures, greatly reducing the discomfort and distress of the patient. A simple and effective protocol for reprogramming blood cells using a constitutive single excisable vector may accelerate the application of iPSC technology by making it accessible to a broader research community. Furthermore, reprogramming of peripheral blood cells allows for the generation of iPSCs from individuals in which skin biopsies should be avoided (i.e. aberrant scarring) or due to pre-existing disease conditions preventing access to punch biopsies. Here we demonstrate a protocol for the generation of human iPSCs from peripheral blood mononuclear cells (PBMCs) using a single floxed-excisable lentiviral vector constitutively expressing the 4 factors. Freshly collected or thawed PBMCs are expanded for 9 days as described(10,11) in medium containing ascorbic acid, SCF, IGF-1, IL-3 and EPO before being transduced with the STEMCCA lentivirus. Cells are then plated onto MEFs and ESC-like colonies can be visualized two weeks after infection. Finally, selected clones are expanded and tested for the expression of the pluripotency markers SSEA-4, Tra-1-60 and Tra-1-81. This protocol is simple, robust and highly consistent, providing a reliable methodology for the generation of human iPSCs from readily accessible 4 ml of blood.


Asunto(s)
Lentivirus/genética , Leucocitos Mononucleares/fisiología , Células Madre Pluripotentes/fisiología , Antígenos de Superficie/biosíntesis , Vectores Genéticos/genética , Humanos , Factor 4 Similar a Kruppel , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteoglicanos/biosíntesis , Antígenos Embrionarios Específico de Estadio/biosíntesis , Transducción Genética
8.
Biotechnol Lett ; 34(12): 2307-15, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22983716

RESUMEN

The commercialisation of human embryonic stem cell derived cell therapies for large patient populations is reliant on both minimising expensive and variable manual-handling methods whilst realising economies of scale. The Quantum Cell Expansion System, a hollow fibre bioreactor (Terumo BCT), was used in a pilot study to expand 60 million human embryonic stem cells to 708 million cells. Further improvements can be expected with optimisation of media flow rates throughout the run to better control the cellular microenvironment. High levels of pluripotency marker expression were maintained on the bioreactor, with 97.7 % of cells expressing SSEA-4 when harvested.


Asunto(s)
Reactores Biológicos , Técnicas de Cultivo de Célula/métodos , Células Madre Embrionarias/fisiología , Expresión Génica , Humanos , Antígenos Embrionarios Específico de Estadio/biosíntesis
9.
Differentiation ; 84(4): 330-43, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22906706

RESUMEN

The heterogeneous nature of stem cells is an important issue in both research and therapeutic use in terms of directing cell lineage differentiation pathways, as well as self-renewal properties. Using flow cytometry we have identified two distinct subpopulations by size, large and small, within cultures of human embryonic stem (hES) cell lines. These two cell populations respond differentially to retinoic acid (RA) differentiation and several endocrine disruptor compounds (EDC). The large cell population responds to retinoic acid differentiation with greater than a 50% reduction in cell number and loss of Oct-4 expression, whereas the number of the small cell population does not change and Oct-4 protein expression is maintained. In addition, four estrogenic compounds altered SSEA-3 expression differentially between the two cell subpopulations changing their ratios relative to each other. Both populations express stem cell markers Oct-4, Nanog, Tra-1-60, Tra-1-80 and SSEA-4, but express low levels of differentiation markers common to the three germ layers. Cloning studies indicate that both populations can revive the parental population. Furthermore, whole genome microarray identified approximately 400 genes with significantly different expression between the two populations (p<0.01). We propose the differential response to RA in these populations is due to differential gene expression of Notch signaling members, CoupTF1 and CoupTF2, chromatin remodeling and histone modifying genes that render the small population resistant to RA differentiation. The findings that hES cells exist as heterogeneous populations with distinct responses to differentiation signals and environmental stimuli will be relevant for their use for drug discovery and disease therapy.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Disruptores Endocrinos/farmacología , Tretinoina/farmacología , Antígenos de Diferenciación/metabolismo , Antígenos de Superficie/biosíntesis , Antígenos de Carbohidratos Asociados a Tumores/biosíntesis , Apigenina/farmacología , Factor de Transcripción COUP II/biosíntesis , Ciclo Celular/efectos de los fármacos , Línea Celular , Clordecona/farmacología , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Humanos , Quempferoles/farmacología , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteoglicanos/biosíntesis , Transducción de Señal/efectos de los fármacos , Antígenos Embrionarios Específico de Estadio/biosíntesis , Tamoxifeno/farmacología
10.
In Vitro Cell Dev Biol Anim ; 48(6): 349-58, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22678753

RESUMEN

Embryonic stem cells (ESCs) derived from inner cell mass (ICM) of mammalian blastocyst are having indefinite proliferation and differentiation capability for any type of cell lineages. In the present study, ICMs of in vitro-derived buffalo blastocysts were cultured into two different culture systems using buffalo fetal fibroblast as somatic cell support and Matrigel as synthetic support to obtain pluripotent buffalo embryonic stem cell (buESC) colonies. Pluripotency of the ESCs were characterised through pluripotency markers whereas, their differentiation capability was assessed by teratoma assay using immuno-compromised mice. Cumulus ooccyte complexes from slaughter house-derived ovaries were subjected to in vitro maturation, in vitro fertilization and in vitro culture to generate blastocysts. Total 262 blastocysts were derived through IVEP with 11.83 % (31/262) hatching rate. To generate buESCs, 15 ICMs from hatched blastocysts were cultured on mitomycin-C-treated homologous fetal fibroblast feeder layer, whereas the leftover 16 ICMs were cultured on extra-cellular matrix (Matrigel). No significant differences were observed for primary ESCs colony formation between two culture systems. Primary colonies as well as passaged ESCs were characterised by alkaline phosphatase staining, karyotyping and expression of transcription-based stem cell markers, OCT-4 and cell surface antigens SSEA-4 and TRA-1-60. Batch of ESCs found positive for pluripotency markers and showing normal karyotype after fifteenth passage were inoculated into eight immuno-compromised mice through subcutaneous and intramuscular route. Subcutaneous route of inoculation was found to be better than intramuscular route. Developed teratomas were excised surgically and subjected to histological analysis. Histological findings revealed presence of all the three germinal layer derivatives in teratoma sections. Presence of germinal layer derivatives were further confirmed by reverse transcriptase-polymerase chain reaction for the presence of differentiation markers like nerve cell adhesion molecule, fetal liver kinase-1 and alpha-feto protein for ectoderm, mesoderm and endoderm, respectively.


Asunto(s)
Búfalos/embriología , Diferenciación Celular , Técnicas de Cultivo de Embriones , Células Madre Embrionarias , Animales , Antígenos de Superficie/biosíntesis , Blastocisto , Búfalos/genética , Técnicas de Cultivo de Célula , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/fisiología , Cariotipo , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Oocitos/fisiología , Células Madre Pluripotentes/citología , Antígenos Embrionarios Específico de Estadio/biosíntesis , Teratoma
11.
Cells Tissues Organs ; 196(2): 107-16, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22237379

RESUMEN

We assessed human mesenchymal stem cells (MSCs) harvested from breast and abdominal adipose tissues enriched in embryonic stage-specific antigen (SSEA-4) expression for osteogenic and adipogenic differentiation in comparison to a mixed cell population. Human adipose was obtained from abdominal and breast tissues of females undergoing gastric bypass and breast reduction, respectively. SSEA-4-expressing cells were enriched from the mixed cell population by magnetic cell sorting and expanded in culture. The results showed that freshly isolated cells from breast and abdominal tissues based on adipose from 3 patients comprised 12 and 10% SSEA-4+ cells, respectively. At passage 0, 48% of the cells from breast adipose tissue were positive for SSEA-4 while 12% of the cells from abdominal adipose tissue were positive for this antigen. The level of SSEA-4-expressing cells remained relatively constant with passaging; SSEA-4-expressing cells from breast tissue comprised 45% of the total while 27% of the cells from abdominal adipose tissue expressed SSEA-4 at passage 5. Cells sorted for SSEA-4 expression exhibited a higher potential for differentiation toward osteogenic and adipogenc cell lineages in vitro when compared to a mixed population. Interestingly, SSEA-4 expression was lost upon differentiation, suggesting that the antigen marks a subpopulation of MSCs. Taken together, the data demonstrate that breast adipose tissue is highly enriched in a subpopulation of MSCs expressing SSEA-4 and suggest that SSEA-4 may be a marker of a subpopulation of MSCs with high potential for osteogenic and adipogenic differentiation.


Asunto(s)
Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Mama/citología , Mama/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Antígenos Embrionarios Específico de Estadio/biosíntesis , Anciano , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Femenino , Perfilación de la Expresión Génica , Humanos , Persona de Mediana Edad
12.
Int J Dev Biol ; 55(3): 305-11, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21710436

RESUMEN

Human induced pluripotent stem (hiPS) cells have great potential for regenerative medicine and drug discovery. It is essential to establish highly efficient and reliable methods for hiPS cell cryopreservation. We examined cryopreservation of hiPS cells by the vitrification method using a dimethyl sulfoxide Me2SO-free and serum-free medium, VS2E, that uses Euro-Collins solution as a base with 40% (v/v) ethylene glycol and 10% (w/v) polyethylene glycol as cryoprotectants. This combination of vitrification and cryoprotectants resulted in a higher recovery rate of hiPS cells than with a commercially-available vitrification solution, DAP213, which contained Me2SO and serum components. After vitrification and warming, hiPS cells were cultured easily. Even after several subculturing steps, cells expressed undifferentiated cell markers, such as Oct-3/4 and SSEA-4, and also exhibited alkaline phosphatase activity. The pluripotency of hiPS cells was maintained, as demonstrated by teratoma formation upon hiPS cell transplantation into severe combined immunodeficient mice. Thus, we successfully preserved hiPS cells under liquid nitrogen with high efficiency using Me2SO-free vitrification solution and rapid cooling.


Asunto(s)
Criopreservación/métodos , Crioprotectores/farmacología , Células Madre Pluripotentes Inducidas/metabolismo , Acetamidas , Fosfatasa Alcalina/biosíntesis , Fosfatasa Alcalina/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Dimetilsulfóxido/farmacología , Glicol de Etileno/farmacología , Humanos , Soluciones Hipertónicas/farmacología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ratones , Ratones SCID , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Polietilenglicoles/farmacología , Glicoles de Propileno , Antígenos Embrionarios Específico de Estadio/biosíntesis
13.
PLoS One ; 6(4): e19114, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21559451

RESUMEN

Nocodazole is a known destabiliser of microtubule dynamics and arrests cell-cycle at the G2/M phase. In the context of the human embryonic stem cell (hESC) it is important to understand how this arrest influences the pluripotency of cells. Here we report for the first time the changes in the expression of transcription markers Nanog and Oct4 as well as SSEA-3 and SSEA-4 in human embryonic cells after their treatment with nocodazole. Multivariate permeabilised-cell flow cytometry was applied for characterising the expression of Nanog and Oct4 during different cell cycle phases. Among untreated hESC we detected Nanog-expressing cells, which also expressed Oct4, SSEA-3 and SSEA-4. We also found another population expressing SSEA-4, but without Nanog, Oct4 and SSEA-3 expression. Nocodazole treatment resulted in a decrease of cell population positive for all four markers Nanog, Oct4, SSEA-3, SSEA-4. Nocodazole-mediated cell-cycle arrest was accompanied by higher rate of apoptosis and upregulation of p53. Twenty-four hours after the release from nocodazole block, the cell cycle of hESC normalised, but no increase in the expression of transcription markers Nanog and Oct4 was detected. In addition, the presence of ROCK-2 inhibitor Y-27632 in the medium had no effect on increasing the expression of pluripotency markers Nanog and Oct4 or decreasing apoptosis or the level of p53. The expression of SSEA-3 and SSEA-4 increased in Nanog-positive cells after wash-out of nocodazole in the presence and in the absence of Y-27632. Our data show that in hESC nocodazole reversible blocks cell cycle, which is accompanied by irreversible loss of expression of pluripotency markers Nanog and Oct4.


Asunto(s)
Antineoplásicos/farmacología , Células Madre Embrionarias/citología , Proteínas de Homeodominio/biosíntesis , Nocodazol/uso terapéutico , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Animales , Antígenos de Carbohidratos Asociados a Tumores/biosíntesis , Apoptosis , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Citometría de Flujo/métodos , Humanos , Ratones , Mitosis , Proteína Homeótica Nanog , Antígenos Embrionarios Específico de Estadio/biosíntesis
14.
Stem Cells Dev ; 20(8): 1451-64, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21291304

RESUMEN

The present study was undertaken to detect, characterize, and study differentiation potential of stem cells in adult rabbit, sheep, monkey, and menopausal human ovarian surface epithelium (OSE). Two distinct populations of putative stem cells (PSCs) of variable size were detected in scraped OSE, one being smaller and other similar in size to the surrounding red blood cells in the scraped OSE. The smaller 1-3 µm very small embryonic-like PSCs were pluripotent in nature with nuclear Oct-4 and cell surface SSEA-4, whereas the bigger 4-7 µm cells with cytoplasmic localization of Oct-4 and minimal expression of SSEA-4 were possibly the tissue committed progenitor stem cells. Pluripotent gene transcripts of Oct-4, Oct-4A, Nanog, Sox-2, TERT, and Stat-3 in human and sheep OSE were detected by reverse transcriptase-polymerase chain reaction. The PSCs underwent spontaneous differentiation into oocyte-like structures, parthenote-like structures, embryoid body-like structures, cells with neuronal-like phenotype, and embryonic stem cell-like colonies, whereas the epithelial cells transformed into mesenchymal phenotype by epithelial-mesenchymal transition in 3 weeks of OSE culture. Germ cell markers like c-Kit, DAZL, GDF-9, VASA, and ZP4 were immuno-localized in oocyte-like structures. In conclusion, as opposed to the existing view of OSE being a bipotent source of oocytes and granulosa cells, mammalian ovaries harbor distinct very small embryonic-like PSCs and tissue committed progenitor stem cells population that have the potential to develop into oocyte-like structures in vitro, whereas mesenchymal fibroblasts appear to form supporting granulosa-like somatic cells. Research at the single-cell level, including complete gene expression profiling, is required to further confirm whether postnatal oogenesis is a conserved phenomenon in adult mammals.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Oogénesis/fisiología , Ovario/citología , Adulto , Animales , Técnicas de Cultivo de Célula , Proliferación Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Femenino , Haplorrinos , Humanos , Menopausia , Persona de Mediana Edad , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Oocitos/citología , Oocitos/metabolismo , Ovario/metabolismo , Conejos , Ovinos , Antígenos Embrionarios Específico de Estadio/biosíntesis
15.
Int J Dev Biol ; 55(2): 181-7, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21305471

RESUMEN

Human bone marrow-derived mesenchymal stem cells (hMSCs) are potential cellular sources of therapeutic stem cells as they have the ability to proliferate and differentiate into a wide array of mesenchymal cell types such as osteoblasts, chondroblasts and adipocytes. hMSCs have been used clinically to treat patients with graft vs. host disease, osteogenesis imperfect, or alveolar cleft, suggesting that transplantation of hMSCs is comparatively safe as a stem cell-based therapy. However, conventional culture medium for hMSCs contains fetal bovine serum (FBS). In the present study, we developed a growth factor-defined, serum-free medium for culturing hMSCs. Under these conditions, TGF-beta1 promoted proliferation of hMSCs. The expanded hMSC population expressed the human pluripotency markers SSEA-3, -4, NANOG, OCT3/4 and SOX2. Furthermore, double positive cells for SSEA-3 and a mesenchymal cell marker, CD105, were detected in the population. The potential to differentiate into osteoblasts and adipocytes was confirmed. This work provides a useful tool to understand the basic biological properties of hMSCs in culture.


Asunto(s)
Proliferación Celular , Péptidos y Proteínas de Señalización Intercelular/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Pluripotentes/metabolismo , Técnicas de Cultivo de Tejidos , Antígenos CD/biosíntesis , Antígenos de Carbohidratos Asociados a Tumores/biosíntesis , Diferenciación Celular , Células Cultivadas , Medio de Cultivo Libre de Suero , Endoglina , Proteínas de Homeodominio/biosíntesis , Humanos , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Receptores de Superficie Celular/biosíntesis , Factores de Transcripción SOXB1/biosíntesis , Antígenos Embrionarios Específico de Estadio/biosíntesis , Factor de Crecimiento Transformador beta/farmacología
16.
J Dent Res ; 90(3): 317-24, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21076121

RESUMEN

UNLABELLED: Although human orofacial bone-marrow-derived mesenchymal stem cells showed differentiation traits distinctly different from those of mesenchymal stem cells (MSCs) derived from long bone marrow (BMMSCs), mouse MSCs derived from orofacial bone have not been isolated due to technical difficulties, which in turn precludes the use of mouse models to study and cure orofacial diseases. In this study, we developed techniques to isolate and expand mouse orofacial bone/bone-marrow-derived MSCs (OMSCs) from mandibles and verified their MSC characteristics by single-colony formation, multi-lineage differentiation, and in vivo tissue regeneration. Activated T-lymphocytes impaired OMSCs via the Fas/Fas ligand pathway, as occurs in BMMSCs. Furthermore, we found that OMSCs are distinct from BMMSCs with respect to regulating T-lymphocyte survival and proliferation. Analysis of our data suggests that OMSCs are a unique population of MSCs and play an important role in systemic immunity. ABBREVIATIONS: BMMSC, bone marrow mesenchymal stem cell; HA/TCP, hydroxyapatite/tricalcium phosphate; OMSC, orofacial mesenchymal stem cell; OVX, ovariectomized.


Asunto(s)
Mandíbula/citología , Células Madre Mesenquimatosas/citología , Animales , Antígenos Ly/biosíntesis , Apoptosis , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Fosfatos de Calcio , Técnicas de Cultivo de Célula , Diferenciación Celular , Separación Celular , Células Cultivadas , Técnicas de Cocultivo , Ensayo de Unidades Formadoras de Colonias , Durapatita , Proteínas de la Membrana/biosíntesis , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Desnudos , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Osteoblastos/citología , Osteoblastos/metabolismo , Ovariectomía , Regeneración , Antígenos Embrionarios Específico de Estadio/biosíntesis , Linfocitos T/inmunología , Andamios del Tejido
17.
J Vis Exp ; (38)2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20383120

RESUMEN

Human pluripotent stem cells (hPSCs) that include human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) are exciting cell sources due to their limitless self-renewal capabilities and their potential to differentiate into multiple cell types. The pluripotent state of hPSCs is typically assessed by techniques such as qPCR, immunocytochemistry, and by other in vitro and in vivo differentiation strategies into multiple cell types. Among these, immunocytochemical techniques have been developed for routine characterization of the undifferentiated state of hPSCs based on analysis of candidate intracellular and cell-surface biomarkers. Given the fact that hPSCs grow as colonies, problems arise in quantifying the expression of these markers at the individual cell level on a routine basis. Flow cytometry analyses serve to address this issue but require cell numbers and use of reagents that are not normally conducive for routine quality control assessment of hPSC cultures. Thus, the development of practical and reproducible means of creating monolayer cell samples with preserved integrity for marker evaluation has many advantages in stem cell research. This greatly benefits immunocytochemical analysis because individual cells from the monolayer can be easily observed and quantified for the expression of specific markers. Towards this goal, a self-made cytospin apparatus was constructed and optimized for use with immunocytochemical staining. Two cell-surface markers (SSEA3/SSEA4) expression were analyzed in a variant BG01 stem cell line for the purpose of this protocol.


Asunto(s)
Inmunohistoquímica/instrumentación , Inmunohistoquímica/métodos , Células Madre Pluripotentes/metabolismo , Antígenos de Carbohidratos Asociados a Tumores/análisis , Antígenos de Carbohidratos Asociados a Tumores/biosíntesis , Humanos , Células Madre Pluripotentes/química , Células Madre Pluripotentes/citología , Antígenos Embrionarios Específico de Estadio/análisis , Antígenos Embrionarios Específico de Estadio/biosíntesis
18.
Cloning Stem Cells ; 11(3): 427-35, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19751112

RESUMEN

BMP-11/GDF-11 and Myostatin/GDF-8 are both members of the TGF-beta superfamily that can activate SMAD2/3 phosphorylation via the type I receptors ALK4, ALK5, or ALK7. We tested the ability of BMP-11 and Myostatin to promote self-renewal of human embryonic stem cells (hESC) under feeder-free and serum-free culture conditions in short term (1 week) and medium term cultures (10 weeks). We show that hESC cultured in serum-free medium supplemented with either 20 ng/mL Myostatin or 20 ng/mL BMP-11 maintain the colony and cellular morphology of undifferentiated hESC, maintain POU5f1, NANOG, TRA-1-60, and SSEA4 expression, and display increased SMAD2/3 phosphorylation, similar to hESC cultured in mouse embryonic fibroblast feeder-CM or 20 ng/mL Activin-A. The type I TGF-beta receptor inhibitor SB431542 totally inhibited the maintenance activity of both Myostatin or BMP-11 supplemented medium. Our data show that members of the TGF-beta superfamily, other than Activin-A and GDF3, are able to maintain hES cells in an undifferentiated state under feeder free conditions.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Factores de Diferenciación de Crecimiento/farmacología , Miostatina/farmacología , Receptores de Activinas Tipo I/antagonistas & inhibidores , Receptores de Activinas Tipo I/biosíntesis , Activinas/farmacología , Animales , Antígenos de Superficie/biosíntesis , Benzamidas/farmacología , Células Cultivadas , Técnicas de Cocultivo , Dioxoles/farmacología , Proteínas de Homeodominio/biosíntesis , Humanos , Ratones , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteoglicanos/biosíntesis , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Antígenos Embrionarios Específico de Estadio/biosíntesis , Factores de Tiempo
19.
Stem Cell Rev Rep ; 5(1): 72-80, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19184635

RESUMEN

A major concern in human embryonic stem cell (hESC)-derived cell replacement therapy is the risk of tumorigenesis from undifferentiated hESCs residing in the population of hESC-derived cells. Separation of these undifferentiated hESCs from the differentiated derivatives using cell sorting methods may be a plausible approach in overcoming this problem. We therefore explored magnetic activated cell sorting (MACS) and fluorescence activated cell sorting (FACS) to separate labelled undifferentiated hESCs from a heterogeneous population of hESCs and hepatocellular carcinoma cells (HepG2) deliberately mixed respectively at different ratios (10:90, 20:80, 30:70, 40:60 and 50:50) to mimic a standard in vitro differentiation protocol, instead of using a hESC-differentiated cell population, so that we could be sure of the actual number of cells separated. HES-3 and HES-4 cells were labelled in separate experiments for the stem cell markers SSEA-4 and TRA-1-60 using primary antibodies. Anti-PE magnetic microbeads that recognize the PE-conjugated SSEA-4 labelled hESCs was added to the heterogeneous cell mixture and passed through the MACS column. The cells that passed through the column ('flow-through' fraction) and those retained ('labelled' fraction') were subsequently analysed using FACS. The maximum efficacy of hESCs retention using MACS was 81.0 +/- 2.9% (HES-3) and 83.6 +/- 4.2% (HES-4). Using FACS, all the undifferentiated hESCs labelled with the two cell-surface markers could be removed by selective gating. Both hESCs and HepG2 cells in the 'flow-through' fraction following MACS separation were viable in culture whereas by FACS separation only the HepG2 cells were viable. FACS efficiently helps to eliminate the undifferentiated hESCs based on their cell-surface antigens expressed.


Asunto(s)
Antígenos de Superficie/biosíntesis , Separación Celular/métodos , Citometría de Flujo/métodos , Proteoglicanos/biosíntesis , Antígenos Embrionarios Específico de Estadio/biosíntesis , Antígenos de Superficie/análisis , Biomarcadores de Tumor , Diferenciación Celular , Línea Celular Tumoral , Técnicas Citológicas , Humanos , Magnetismo , Coloración y Etiquetado
20.
Stem Cells ; 26(12): 3068-74, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18787205

RESUMEN

OCT4 is a master regulator of self-renewal in embryonic stem cells and can potentially encode two spliced variants, designated OCT4A and OCT4B. We have examined the expression pattern of these OCT4 isoforms in various human pluripotent and nonpluripotent cells. Our data revealed that whereas OCT4A expression is restricted to embryonic stem (ES) and embryonal carcinoma (EC) cells, OCT4B can be detected in various nonpluripotent cell types. Furthermore, we detected a novel OCT4 spliced variant, designated OCT4B1, that is expressed primarily in human ES and EC cells and is downregulated following their differentiation. We also found a significantly higher level of OCT4B1 expression in stage-specific embryonic antigen-3 (SSEA3)(+) compared with SSEA3(-) subpopulations of cultured ES cells. Taken together, our data demonstrated a distinctive expression pattern for OCT4 spliced variants in different cell types and highlight the necessity of defining the type of OCT4 when addressing the expression of this gene in different human cells.


Asunto(s)
Empalme Alternativo , Regulación de la Expresión Génica , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/metabolismo , Células Madre/metabolismo , Antígenos de Carbohidratos Asociados a Tumores/biosíntesis , Diferenciación Celular , Línea Celular Tumoral , Células Madre Embrionarias/citología , Exones , Células HeLa , Humanos , Intrones , Células K562 , Isoformas de Proteínas , Antígenos Embrionarios Específico de Estadio/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...