Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Stem Cells Transl Med ; 13(5): 415-424, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38513284

RESUMO

BACKGROUND: Surgical intervention is the main therapy for refractory vitiligo. We developed a modified autologous cultured epithelial grafting (ACEG) technique for vitiligo treatment. Between January 2015 and June 2019, a total of 726 patients with vitiligo underwent ACEG in China, with patient characteristics and clinical factors being meticulously documented. Using a generalized linear mixed model, we were able to assess the association between these characteristics and the repigmentation rate. RESULTS: ACEG demonstrated a total efficacy rate of 82.81% (1754/2118) in treating 726 patients, with a higher repigmentation rate of 64.87% compared to conventional surgery at 52.69%. Notably, ACEG showed a better response in treating segmental vitiligo, lesions on lower limbs, age ≤ 18, and stable period > 3 years. A keratinocyte:melanocyte ratio below 25 was found to be advantageous too. Single-cell RNA sequencing analysis revealed an increase in melanocyte count and 2 subclusters of keratinocytes after ACEG, which remained higher in repigmented sites even after 1 year. CONCLUSIONS: ACEG is a promising therapy for refractory vitiligo. Patient age, clinical type, lesion site, and stability before surgery influence repigmentation in ACEG. The mechanism of repigmentation after ACEG treatment is likely not confined to the restoration of melanocyte populations. It may also involve an increase in the number of keratinocytes that support melanocyte function within the affected area. These keratinocytes may aid the post-transplant survival and function of melanocytes by secreting cytokines and extracellular matrix components. TRIAL REGISTRATION: registered with Chictr.org.cn (ChiCTR2100051405).


Assuntos
Transplante Autólogo , Vitiligo , Humanos , Vitiligo/terapia , Masculino , Feminino , Estudos Retrospectivos , Transplante Autólogo/métodos , Adulto , Adolescente , Adulto Jovem , Pessoa de Meia-Idade , Melanócitos/transplante , Criança , Queratinócitos/transplante , Células Cultivadas , Epitélio
2.
Dermatol Surg ; 45(4): 497-505, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30096104

RESUMO

BACKGROUND: Autologous cultured therapy has emerged as an effective treatment for stable vitiligo. However, culture methods may include harmful agents and be unsuitable for therapeutic use in humans. OBJECTIVE: To investigate the safety and efficacy of autologous cultured epithelial sheets propagated under serum-free and feeder-free conditions for the treatment of stable vitiligo. METHODS: Twenty-eight patients with stable vitiligo were included in this study. Keratinocytes and melanocytes from 14 patients were cultured under serum-free, feeder-free conditions (Group A). Epithelial cells from the remaining 14 patients were cultured according to Rheinward and Green's technique (Group B). Patients were followed up at 1, 3, 6, and 12 months after transplantation. RESULTS: The epithelial sheets cultured in Group A were thinner and more fragile than Group B, but there were no significant differences in repigmentation between the 2 groups. At 12-month follow-up, in Group A, repigmentation at graft sites was classified as excellent in 9 patients and good in 2 patients. In Group B, repigmentation was excellent in 8 patients and good in 4 patients. Scars at the donor sites were the most frequent adverse events associated with the procedure. CONCLUSION: Autologous epithelial sheet cultured in serum-free, feeder-free conditions is a safe and efficacious approach to cure stable vitiligo.


Assuntos
Técnicas de Cultura de Células/métodos , Células Epiteliais/transplante , Queratinócitos/transplante , Melanócitos/transplante , Transplante de Pele/métodos , Pele/citologia , Vitiligo/cirurgia , Adolescente , Adulto , Criança , Feminino , Humanos , Queratinócitos/citologia , Masculino , Melanócitos/citologia , Pessoa de Meia-Idade , Engenharia Tecidual , Transplante Autólogo , Adulto Jovem
3.
Lab Invest ; 91(4): 553-64, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21283079

RESUMO

Few reports have examined the effects of adult bone marrow multipotent stromal cells (MSCs) on large animals, and no useful method has been established for MSC implantation. In this study, we investigate the effects of MSC infusion from the coronary vein in a swine model of chronic myocardial infarction (MI). MI was induced in domestic swine by placing beads in the left coronary artery. Bone marrow cells were aspirated and then cultured to isolate the MSCs. At 4 weeks after MI, MSCs labeled with dye (n=8) or vehicle (n=5) were infused retrogradely from the anterior interventricular vein without any complications. Left ventriculography (LVG) was performed just before and at 4 weeks after cell infusion. The ejection fraction (EF) assessed by LVG significantly decreased from baseline up to a follow-up at 4 weeks in the control group (P<0.05), whereas the cardiac function was preserved in the MSC group. The difference in the EF between baseline and follow-up was significantly greater in the MSC group than in the control group (P<0.05). The MSC administration significantly promoted neovascularization in the border areas compared with the controls (P<0.0005), though it had no affect on cardiac fibrosis. A few MSCs expressed von Willebrand factor in a differentiation assay, but none of them expressed troponin T. In quantitative gene expression analysis, basic fibroblast growth factor and vascular endothelial growth factor (VEGF) levels were significantly higher in the MSC-treated hearts than in the controls (P<0.05, respectively). Immunohistochemical staining revealed VEGF production in the engrafted MSCs. In vitro experiment demonstrated that MSCs significantly stimulated endothelial capillary network formation compared with the VEGF protein (P<0.0001). MSC infusion via the coronary vein prevented the progression of cardiac dysfunction in chronic MI. This favorable effect appeared to derive not from cell differentiation, but from enhanced neovascularization by angiogenic factors secreted from the MSCs.


Assuntos
Transplante de Medula Óssea/métodos , Coração/fisiopatologia , Células-Tronco Multipotentes/transplante , Isquemia Miocárdica/fisiopatologia , Isquemia Miocárdica/cirurgia , Neovascularização Fisiológica , Células Estromais/transplante , Animais , Diferenciação Celular , Doença Crônica , Vasos Coronários , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fibrose , Infusões Intravenosas , Células-Tronco Multipotentes/metabolismo , Células-Tronco Multipotentes/patologia , Infarto do Miocárdio/complicações , Isquemia Miocárdica/etiologia , Isquemia Miocárdica/patologia , Miocárdio/patologia , Fenótipo , Células Estromais/metabolismo , Células Estromais/patologia , Suínos , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
J Cell Physiol ; 221(2): 335-42, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19582773

RESUMO

Mesenchymal stem cells (MSCs) are attracting a great deal of attention because they represent a valuable source of cells for use in regenerative medicine. In human cell culture it is important to obtain large numbers of cells for use in therapy. In this study, we attempted to prolong life span of a marrow-derived mesenchymal stem cell using a combination of growth factors and hormones. Furthermore we tested whether chemically defined culture conditions are sufficient for maintenance of multipotent mesenchymal stem cells. Epidermal growth factor, platelet-derived growth factor-BB (PDGF-BB), acidic fibroblast growth factor (FGF), basic FGF, and leukemia inhibitory factor were found to be key factors for the mesenchymal stem cell proliferation. The combination of these growth factors showed extremely strong mitogenic activity, and simultaneously induced the expression of cyclin-dependent kinase inhibitor p16ink4a protein and premature senescence more rapidly than serum-supported culture conditions. The induction of p16ink4a by growth factors was mediated through the mitogen-activated protein kinase (MAPK) cascade. Excess growth stimulation by growth factors was thus one of the culture stress signals and a trigger of premature senescence at least in human cells.


Assuntos
Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Adulto , Becaplermina , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Meios de Cultura , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-sis
5.
Exp Cell Res ; 315(16): 2727-40, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19559696

RESUMO

POU5F1 (more commonly known as OCT4/3) is one of the stem cell markers, and affects direction of differentiation in embryonic stem cells. To investigate whether cells of mesenchymal origin acquire embryonic phenotypes, we generated human cells of mesodermal origin with overexpression of the chimeric OCT4/3 gene with physiological co-activator EWS (product of the EWSR1 gene), which is driven by the potent EWS promoter by translocation. The cells expressed embryonic stem cell genes such as NANOG, lost mesenchymal phenotypes, and exhibited embryonal stem cell-like alveolar structures when implanted into the subcutaneous tissue of immunodeficient mice. Hierarchical analysis by microchip analysis and cell surface analysis revealed that the cells are subcategorized into the group of human embryonic stem cells and embryonal carcinoma cells. These results imply that cells of mesenchymal origin can be traced back to cells of embryonic phenotype by the OCT4/3 gene in collaboration with the potent cis-regulatory element and the fused co-activator. The cells generated in this study with overexpression of chimeric OCT4/3 provide us with insight into cell plasticity involving OCT4/3 that is essential for embryonic cell maintenance, and the complexity required for changing cellular identity.


Assuntos
Embrião de Mamíferos , Mesoderma/fisiologia , Morfogênese/fisiologia , Fator 3 de Transcrição de Octâmero , Proteína EWS de Ligação a RNA/metabolismo , Proteínas Recombinantes de Fusão , Animais , Biomarcadores/metabolismo , Linhagem da Célula , Células Cultivadas , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Cariotipagem , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Análise de Componente Principal , Proteína EWS de Ligação a RNA/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Teratoma/metabolismo , Teratoma/patologia
6.
Stem Cells ; 26(7): 1695-704, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18420831

RESUMO

Stem cell therapy can help repair damaged heart tissue. Yet many of the suitable cells currently identified for human use are difficult to obtain and involve invasive procedures. In our search for novel stem cells with a higher cardiomyogenic potential than those available from bone marrow, we discovered that potent cardiac precursor-like cells can be harvested from human menstrual blood. This represents a new, noninvasive, and potent source of cardiac stem cell therapeutic material. We demonstrate that menstrual blood-derived mesenchymal cells (MMCs) began beating spontaneously after induction, exhibiting cardiomyocyte-specific action potentials. Cardiac troponin-I-positive cardiomyocytes accounted for 27%-32% of the MMCs in vitro. The MMCs proliferated, on average, 28 generations without affecting cardiomyogenic transdifferentiation ability, and expressed mRNA of GATA-4 before cardiomyogenic induction. Hypothesizing that the majority of cardiomyogenic cells in MMCs originated from detached uterine endometrial glands, we established monoclonal endometrial gland-derived mesenchymal cells (EMCs), 76%-97% of which transdifferentiated into cardiac cells in vitro. Both EMCs and MMCs were positive for CD29, CD105 and negative for CD34, CD45. EMCs engrafted onto a recipient's heart using a novel 3-dimensional EMC cell sheet manipulation transdifferentiated into cardiac tissue layer in vivo. Transplanted MMCs also significantly restored impaired cardiac function, decreasing the myocardial infarction (MI) area in the nude rat model, with tissue of MMC-derived cardiomyocytes observed in the MI area in vivo. Thus, MMCs appear to be a potential novel, easily accessible source of material for cardiac stem cell-based therapy.


Assuntos
Técnicas de Cultura de Células/métodos , Menstruação/sangue , Células-Tronco Mesenquimais/citologia , Miócitos Cardíacos/citologia , Adulto , Animais , Células da Medula Óssea/citologia , Técnicas de Cocultura , Feminino , Coração/fisiologia , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , Ratos , Ratos Endogâmicos F344 , Células-Tronco/citologia
7.
Am J Physiol Gastrointest Liver Physiol ; 293(5): G1089-98, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17884977

RESUMO

Human umbilical cord blood-derived mesenchymal stem cells (UCBMSCs) are expected to be an excellent source of cells for transplantation. In addition, the stem cell plasticity of human UCBMSCs, which can transdifferentiate into hepatocytes, has been reported. However, the mechanisms involved remain to be clarified. To identify the genes and/or signals that are important in specifying the hepatic fate of human UCBMSCs, we analyzed gene expression profiles during the hepatic differentiation of UCBMSCs with human telomerase reverse transcriptase, UCBMSCs immortalized by infection with a retrovirus carrying telomerase reverse transcriptase, but whose differentiation potential remains unchanged. Efficient differentiation was induced by 5-azacytidine (5-aza)/hepatocyte growth factor (HGF)/oncostatin M (OSM)/fibroblast growth factor 2 (FGF2) treatment in terms of function as well as protein expression: 2.5-fold increase in albumin, 4-fold increase in CCAAT enhancer-binding protein alpha, 1.5-fold increase in cytochrome p450 1A1/2, and 8-fold increase in periodic acid-Schiff staining. Consequently, we found that the expression of Wnt/beta-catenin-related genes downregulated, and the translocation of beta-catenin was observed along the cell membrane and in the cytoplasm, although some beta-catenin was still in the nucleus. Downregulation of Wnt/beta-catenin signals in the cells by Fz8-small interference RNA treatment, which was analyzed with a Tcf4 promoter-luciferase assay, resulted in similar hepatic differentiation to that observed with 5-azacytidine/HGF/OSM/FGF2. In addition, the subcellular distribution of beta-catenin was similar to that of cells treated with 5-azacytidine/HGF/OSM/FGF2. In conclusion, the suppression of Wnt/beta-catenin signaling induced the hepatic differentiation of UCBMSCs, suggesting that Wnt/beta-catenin signals play an important role in the hepatic fate specification of human UCBMSCs.


Assuntos
Diferenciação Celular/fisiologia , Sangue Fetal/fisiologia , Fígado/citologia , Fígado/fisiologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Proteínas Wnt/fisiologia , beta Catenina/fisiologia , Primers do DNA , Sangue Fetal/citologia , Humanos , Recém-Nascido , Reação em Cadeia da Polimerase , Transdução de Sinais , Ureia/análise
8.
Hepatol Res ; 37(12): 1068-79, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17627621

RESUMO

AIM: Bone marrow-derived mesenchymal stem cells (MSC) are expected to be an excellent source of cells for transplantation. We aimed to study the culture conditions and involved genes to differentiate MSC into hepatocytes. METHODS: The culture conditions to induce the efficient differentiation of human bone marrow-derived UE7T-13 cells were examined using cytokines, hormones, 5-azacytidine and type IV collagen. RESULTS: We found that combination of acidic fibroblast growth factor (aFGF), basic fibroblast growth factor (bFGF) and hepatocyte growth factor (HGF) with type IV collagen coating induced hepatic differentiation of UE7T-13 cells at over 30% frequency, where expression of albumin mRNA was increased over 20-fold. The differentiated cells had functions of albumin production, glycogen synthesis and urea secretion as well as expressing hepatocyte-specific genes. In addition, these cellshave binuclear and cuboidal morphology, which is a characteristic feature of hepatocytes. During hepatic differentiation, UE7T-13 cells showed depressed expression of WISP1 and WISP2 genes, members of the CCN family. Conversely, knockdown of WISP1 or WISP2 gene by siRNA stimulated hepatic differentiation. The effect of aFGF/bFGF/HGF/type IV collagen coating and WISP1-siRNA on hepatic differentiation was additive. CONCLUSION: The present study suggests that aFGF/bFGF/HGF/type IV collagen coating is the efficient condition for hepatic differentiation of UE7T-13 cells, and that WISP1 and WISP2 play an important role in hepatic transdifferentiation of these cells.

9.
Exp Cell Res ; 313(12): 2550-62, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17544394

RESUMO

The clinical application of cell transplantation for severe heart failure is a promising strategy to improve impaired cardiac function. Recently, an array of cell types, including bone marrow cells, endothelial progenitors, mesenchymal stem cells, resident cardiac stem cells, and embryonic stem cells, have become important candidates for cell sources for cardiac repair. In the present study, we focused on the placenta as a cell source. Cells from the chorionic plate in the fetal portion of the human placenta were obtained after delivery by the primary culture method, and the cells generated in this study had the Y sex chromosome, indicating that the cells were derived from the fetus. The cells potentially expressed 'working' cardiomyocyte-specific genes such as cardiac myosin heavy chain 7beta, atrial myosin light chain, cardiac alpha-actin by gene chip analysis, and Csx/Nkx2.5, GATA4 by RT-PCR, cardiac troponin-I and connexin 43 by immunohistochemistry. These cells were able to differentiate into cardiomyocytes. Cardiac troponin-I and connexin 43 displayed a discontinuous pattern of localization at intercellular contact sites after cardiomyogenic differentiation, suggesting that the chorionic mesoderm contained a large number of cells with cardiomyogenic potential. The cells began spontaneously beating 3 days after co-cultivation with murine fetal cardiomyocytes and the frequency of beating cells reached a maximum on day 10. The contraction of the cardiomyocytes was rhythmical and synchronous, suggesting the presence of electrical communication between the cells. Placenta-derived human fetal cells may be useful for patients who cannot supply bone marrow cells but want to receive stem cell-based cardiac therapy.


Assuntos
Potenciais de Ação , Córion/citologia , Mesoderma/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Placenta/citologia , Actinina/metabolismo , Animais , Diferenciação Celular , Conexina 43/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Troponina I/metabolismo
10.
Stem Cells ; 25(8): 2017-24, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17495114

RESUMO

We tested the cardiomyogenic potential of the human umbilical cord blood-derived mesenchymal stem cells (UCBMSCs). Both the number and function of stem cells may be depressed in senile patients with severe coronary risk factors. Therefore, stem cells obtained from such patients may not function well. For this reason, UCBMSCs are potentially a new cell source for stem cell-based therapy, since such cells can be obtained from younger populations and are being routinely utilized for clinical patients. The human UCBMSCs (5 x 10(3) per cm(2)) were cocultured with fetal murine cardiomyocytes ([CM] 1 x 10(5) per cm(2)). On day 5 of cocultivation, approximately half of the green fluorescent protein (GFP)-labeled UCBMSCs contracted rhythmically and synchronously, suggesting the presence of electrical communication between the UCBMSCs. The fractional shortening of the contracted UCBMSCs was 6.5% +/- 0.7% (n = 20). The UCBMSC-derived cardiomyocytes stained positive for cardiac troponin-I (clear striation +) and connexin 43 (diffuse dot-like staining at the margin of the cell) by the immunocytochemical method. Cardiac troponin-I positive cardiomyocytes accounted for 45% +/- 3% of GFP-labeled UCBMSCs. The cardiomyocyte-specific long action potential duration (186 +/- 12 milliseconds) was recorded with a glass microelectrode from the GFP-labeled UCBMSCs. CM were observed in UCBMSCs, which were cocultivated in the same dish with mouse cardiomyocytes separated by a collagen membrane. Cell fusion, therefore, was not a major cause of CM in the UCBMSCs. Approximately half of the human UCBMSCs were successfully transdifferentiated into cardiomyocytes in vitro. UCBMSCs can be a promising cellular source for cardiac stem cell-based therapy. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Diferenciação Celular , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Miócitos Cardíacos/citologia , Animais , Antígenos de Superfície/metabolismo , Biomarcadores/metabolismo , Fusão Celular , Células Cultivadas , Técnicas de Cocultura , Coração/embriologia , Humanos , Camundongos , Contração Miocárdica/fisiologia , Miócitos Cardíacos/metabolismo , Organismos Geneticamente Modificados , Telomerase/genética
11.
Mol Biol Cell ; 18(5): 1586-94, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17314403

RESUMO

Duchenne muscular dystrophy (DMD), the most common lethal genetic disorder in children, is an X-linked recessive muscle disease characterized by the absence of dystrophin at the sarcolemma of muscle fibers. We examined a putative endometrial progenitor obtained from endometrial tissue samples to determine whether these cells repair muscular degeneration in a murine mdx model of DMD. Implanted cells conferred human dystrophin in degenerated muscle of immunodeficient mdx mice. We then examined menstrual blood-derived cells to determine whether primarily cultured nontransformed cells also repair dystrophied muscle. In vivo transfer of menstrual blood-derived cells into dystrophic muscles of immunodeficient mdx mice restored sarcolemmal expression of dystrophin. Labeling of implanted cells with enhanced green fluorescent protein and differential staining of human and murine nuclei suggest that human dystrophin expression is due to cell fusion between host myocytes and implanted cells. In vitro analysis revealed that endometrial progenitor cells and menstrual blood-derived cells can efficiently transdifferentiate into myoblasts/myocytes, fuse to C2C12 murine myoblasts by in vitro coculturing, and start to express dystrophin after fusion. These results demonstrate that the endometrial progenitor cells and menstrual blood-derived cells can transfer dystrophin into dystrophied myocytes through cell fusion and transdifferentiation in vitro and in vivo.


Assuntos
Distrofina/genética , Menstruação/sangue , Distrofia Muscular Animal/terapia , Animais , Diferenciação Celular , Fusão Celular , Linhagem Celular , Endométrio/citologia , Feminino , Técnicas de Transferência de Genes , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos NOD , Camundongos Endogâmicos mdx , Camundongos Knockout , Camundongos SCID , Desenvolvimento Muscular , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia
12.
J Cell Biochem ; 100(5): 1240-54, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17115412

RESUMO

What is it that defines a bone marrow-derived chondrocyte? We attempted to identify marrow-derived cells with chondrogenic nature and immortality without transformation, defining "immortality" simply as indefinite cell division. KUM5 mesenchymal cells, a marrow stromal cell line, generated hyaline cartilage in vivo and exhibited enchondral ossification at a later stage after implantation. Selection of KUM5 chondroblasts based on the activity of the chondrocyte-specific cis-regulatory element of the collagen alpha2(XI) gene resulted in enhancement of their chondrogenic nature. Gene chip analysis revealed that OP9 cells, another marrow stromal cell line, derived from macrophage colony-stimulating factor-deficient osteopetrotic mice and also known to be niche-constituting cells for hematopoietic stem cells expressed chondrocyte-specific or -associated genes such as type II collagen alpha1, Sox9, and cartilage oligomeric matrix protein at an extremely high level, as did KUM5 cells. After cultured OP9 micromasses exposed to TGF-beta3 and BMP2 were implanted in mice, they produced abundant metachromatic matrix with the toluidine blue stain and formed type II collagen-positive hyaline cartilage within 2 weeks in vivo. Hierarchical clustering and principal component analysis based on microarray data of the expression of cell surface markers and cell-type-specific genes resulted in grouping of KUM5 and OP9 cells into the same subcategory of "chondroblast," that is, a distinct cell type group. We here show that these two cell lines exhibit the unique characteristics of hyaline cartilage formation and enchondral ossification in vitro and in vivo.


Assuntos
Condrócitos/citologia , Condrogênese , Lâmina de Crescimento/fisiologia , Cartilagem Hialina/citologia , Osteogênese , Animais , Biomarcadores/metabolismo , Desenvolvimento Ósseo , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/farmacologia , Células Cultivadas , Condrócitos/metabolismo , Colágeno Tipo II , Perfilação da Expressão Gênica , Lâmina de Crescimento/transplante , Cartilagem Hialina/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Células Estromais/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta3/farmacologia
13.
J Lab Clin Med ; 146(5): 271-8, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16242526

RESUMO

It has been shown that the stromal-cell population found in bone marrow can be expanded and differentiated into cells with the phenotypes of bone, cartilage, muscle, neural, and fat cells. However, whether mesenchymal stem cells (MSCs) are present in human umbilical-cord blood (UCB) has been the subject of ongoing debate. In this study, we report on a population of fibroblastlike cells derived from the mononuclear fraction of human UCB with osteogenic and adipogenic potential, as well as the presence of a subset of cells that have been maintained in continuous culture for more than 6 months. These cells were found to express CD29, CD44, CD90, CD95, CD105, CD166, and MHC class, but not CD14, CD34, CD40, CD45, CD80, CD86, CD117, CD152, or MHC class II. We also compared gene expression after gene transfer using lenti- and adenoviral vectors carrying the green fluorescence protein to the MSCs derived from UCB because a reliable gene-delivery system is required to transfer target genes into MSCs, which have attracted attention as potential platforms for the systemic delivery of therapeutic genes. The lentiviral vectors can transduce these cells more efficiently than can adenoviral vectors, and we maintained transgene expression for at least 5 weeks. This is the first report showing that UCB-derived MSCs can express exogenous genes by way of a lentivirus vector. These results demonstrate that human UCB is a source of mesenchymal progenitors and may be used in cell transplantation and a wide range of gene-therapy treatments.


Assuntos
Sangue Fetal/citologia , Expressão Gênica , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Adenoviridae/genética , Adipócitos/citologia , Adipócitos/metabolismo , Antígenos CD/metabolismo , Diferenciação Celular , Proliferação de Células , Separação Celular , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Citometria de Fluxo , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunofenotipagem , Recém-Nascido , Lentivirus/genética , Osteócitos/citologia , Osteócitos/metabolismo , Transgenes
14.
J Biomed Mater Res B Appl Biomater ; 74(1): 511-9, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15906389

RESUMO

This study investigates the separation of two types of marrow stromal cells, KUSA-A1 osteoblasts and H-1/A preadipocytes, by filtration through various porous polymeric membranes. It was found that KUSA-A1 permeates better than H-1/A cells through 12-microm polyurethane foaming membranes. This appears to be due to the relatively smaller cell size of KUSA-A1 cells. In addition, when feed solutions containing suspensions of either cell type or a mixture of the two were used, the permeation ratio was relatively low (< 6%) through polyurethane and surface-modified polyurethane foaming membranes. It was also found that there was some degree of separation between KUSA-A1 and H-1/A cells (separation factor = 1.8) with nylon-net filter membranes, but no separation was obtained when filters made of nonwoven fabrics or silk screens were used. This ability of the nylon-net filter membranes to separate the two cell types was due to a sieving effect that results from an optimal pore size. Finally, permeation of a solution of human serum albumin through the membrane following filtration of the cells did not result in a separation of cells in the recovery solution.


Assuntos
Materiais Biocompatíveis/química , Separação Celular/instrumentação , Separação Celular/métodos , Membranas Artificiais , Mesoderma/patologia , Polímeros/química , Células-Tronco/citologia , Adipócitos/citologia , Animais , Células da Medula Óssea/citologia , Membrana Celular , Citometria de Fluxo , Mesoderma/ultraestrutura , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Varredura , Modelos Químicos , Osteoblastos/metabolismo , Poliuretanos/química , Espalhamento de Radiação , Células-Tronco/ultraestrutura , Células Estromais/citologia , Propriedades de Superfície
15.
Mol Biol Cell ; 16(3): 1491-9, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15647378

RESUMO

Human umbilical cord blood-derived mesenchymal stem cells (UCBMSCs) are expected to serve as an excellent alternative to bone marrow-derived human mesenchymal stem cells. However, it is difficult to study them because of their limited life span. To overcome this problem, we attempted to produce a strain of UCBMSCs with a long life span and to investigate whether the strain could maintain phenotypes in vitro. UCBMSCs were infected with retrovirus carrying the human telomerase reverse transcriptase (hTERT) to prolong their life span. The UCBMSCs underwent 30 population doublings (PDs) and stopped dividing at PD 37. The UCBMSCs newly established with hTERT (UCBTERTs) proliferated for >120 PDs. The p16INK4a/RB braking pathway leading to senescence can be inhibited by introduction of Bmi-1, a polycomb-group gene, and human papillomavirus type 16 E7, but the extension of the life span of the UCBMSCs with hTERT did not require inhibition of the p16INK4a/RB pathway. The characteristics of the UCBTERTs remained unchanged during the prolongation of life span. UCBTERTs provide a powerful model for further study of cellular senescence and for future application to cell-based therapy by using umbilical cord blood cells.


Assuntos
Técnicas de Cultura de Células/métodos , Células Cultivadas/citologia , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Sangue Fetal/citologia , Proteína do Retinoblastoma/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Western Blotting , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Transplante de Células , Senescência Celular , Proteínas de Ligação a DNA , Citometria de Fluxo , Humanos , Cariotipagem , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Osteócitos/citologia , Proteínas E7 de Papillomavirus , Fenótipo , Complexo Repressor Polycomb 1 , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Retroviridae/genética , Células-Tronco/citologia , Telomerase/metabolismo , Telômero/metabolismo , Telômero/ultraestrutura , Fatores de Tempo
16.
Microsc Res Tech ; 56(6): 421-34, 2002 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11921344

RESUMO

Since Henze discovered vanadium in the blood (or coelomic) cells of an ascidian in 1911, this unusual phenomenon has attracted the interest of many investigators. The highest concentration of vanadium (350 mM) in the blood cells of Ascidia gemmata, which belongs to the suborder Phlebobranchia, is 10(7) times higher than that in seawater. Of the approximately 10 types of blood cells, a combination of cell fractionation and neutron-activation analysis revealed that the signet ring cells were the true vanadocytes. In the vanadocytes, 97.6% of the vanadium is in the +3 oxidation state (III). The extremely low pH of 1.9 found in vanadocytes suggests that protons, concentrated by an H(+)-ATPase, might be linked to the accumulation of vanadium energetically. The antigen recognized by a monoclonal antibody, S4D5, prepared to identify vanadocytes, was determined to be 6-PGDH in the pentose phosphate pathway. NADPH produced in the pentose phosphate pathway in vanadocytes is thought to participate in the reduction of vanadium(V) to vanadium(IV). During embryogenesis, a vanadocyte-specific antigen first appears in the body wall at the same time that significant accumulations of vanadium become apparent. Three different vanadium-associated proteins (VAPs) were extracted from the blood cells of vanadium-rich ascidians. These are 12.5, 15, and 16 kDa in size and are associated with vanadium in an approximate ratio of 1:16. The cDNA encoding the 12.5 and 15 kDa VAPs was isolated and the proteins encoded were found to be novel. Further biochemical and biophysical characterization of the VAPs is in progress.


Assuntos
Células Sanguíneas/metabolismo , Urocordados/metabolismo , Vanádio/sangue , Animais , Anticorpos Monoclonais , Oxirredução , ATPases Translocadoras de Prótons/metabolismo , Urocordados/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA