Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Immunol ; 50(1): 110-118, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31706262

RESUMO

Mast cells are generally classified into two phenotypically distinct populations: mucosal-type mast cells (MMCs) and connective tissue-type mast cells (CTMCs). However, the molecular basis determining the different characteristics of the mast cell subclasses still remains unclear. Unfortunately, the number of mast cells that can be obtained from tissues is limited, which makes it difficult to study the function of each mast cell subclass. Here, we report the generation and characterization of MMCs and CTMCs derived from mouse BM mast cells (BMMCs). We found that the expression of galanin receptor 3 was elevated in MMCs when compared to the expression in CTMCs. Moreover, intraperitoneal injection of a galanin antagonist reduced MMCs and inhibited the inflammation of dextran sodium sulfate-induced colitis in mice. Therefore, these results suggest that galanin promotes MMC differentiation in vivo, and provide important insights into the molecular mechanisms underlying the differentiation of mast cell subclasses.


Assuntos
Diferenciação Celular/imunologia , Galanina/metabolismo , Mastócitos/citologia , Mucosa/citologia , Mucosa/imunologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Mastócitos/imunologia , Mastócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
2.
Biomed Res Int ; 2016: 2048987, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27843938

RESUMO

Mast cells play an important role in the pathogenesis of allergic diseases. Immature mast cells migrate into peripheral tissues from the bone marrow and undergo complete maturation. Interestingly, mast cells have characteristics similar to hematopoietic stem cells (HSCs), such as self-renewal and c-kit expression. In HSCs, Wnt signaling is involved in their maintenance and differentiation. On the other hand, the relation between Wnt signaling and mast cell differentiation is poorly understood. To study whether Wnt signals play a role in the maturation of mast cells, we studied the effect of Wnt proteins on mast cell maturation of bone marrow-derived mast cells (BMMCs). The expression levels of CD81 protein and histidine decarboxylase mRNA and activity of mast cell-specific protease were all elevated in BMMCs treated with Wnt5a. In addition, Wnt5a induced the expression of Axin2 and TCF mRNA in BMMCs. These results showed that Wnt5a could promote the maturation of mast cells via the canonical Wnt signaling pathway and provide important insights into the molecular mechanisms underlying the differentiation of mast cells.


Assuntos
Diferenciação Celular/genética , Hipersensibilidade/genética , Mastócitos/metabolismo , Proteína Wnt-5a/genética , Animais , Proteína Axina/biossíntese , Células da Medula Óssea/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Histidina Descarboxilase/biossíntese , Hipersensibilidade/patologia , Mastócitos/citologia , Camundongos , Tetraspanina 28/biossíntese , Via de Sinalização Wnt/genética , Proteína Wnt-5a/administração & dosagem , Proteína Wnt-5a/metabolismo
3.
Methods Mol Biol ; 1341: 181-95, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26138986

RESUMO

Flk1-expressing (+) mesodermal cells are useful source for the generation of hematopoietic cells and cardiomyocytes from pluripotent stem cells (PSCs). However, they have been reported as a heterogenous population that includes hematopoietic and cardiac progenitors. Therefore, to provide a method for a highly efficient production of hematopoietic cells and cardiomyocytes, cell surface markers are often used for separating these progenitors in Flk1(+) cells. Our recent study has shown that the expression of coxsackievirus and adenovirus receptor (CAR), a tight junction component molecule, could divide mouse and human PSC- and mouse embryo-derived Flk1(+) cells into Flk1(+)CAR(-) and Flk1(+)CAR(+) cells. Flk1(+)CAR(-) and Flk1(+)CAR(+) cells efficiently differentiated into hematopoietic cells and cardiomyocytes, respectively. These results indicate that CAR is a novel cell surface marker for separating PSC-derived Flk1(+) mesodermal cells into hematopoietic and cardiac progenitors. We herein describe a differentiation method from PSCs into hematopoietic cells and cardiomyocytes based on CAR expression.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células-Tronco Hematopoéticas/citologia , Miócitos Cardíacos/citologia , Células-Tronco Pluripotentes/citologia , Animais , Linhagem Celular , Células Cultivadas , Humanos , Mesoderma/citologia , Camundongos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/análise
4.
PLoS One ; 10(6): e0128890, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26061227

RESUMO

The blood brain barrier (BBB) is formed by brain microvascular endothelial cells (BMECs) and tightly regulates the transport of molecules from blood to neural tissues. In vitro BBB models from human pluripotent stem cell (PSCs)-derived BMECs would be useful not only for the research on the BBB development and function but also for drug-screening for neurological diseases. However, little is known about the differentiation of human PSCs to BMECs. In the present study, human induced PSCs (iPSCs) were differentiated into endothelial cells (ECs), and further maturated to BMECs. Interestingly, C6 rat glioma cell-conditioned medium (C6CM), in addition to C6 co-culture, induced the differentiation of human iPSC-derived ECs (iPS-ECs) to BMEC-like cells, increase in the trans-endothelial electrical resistance, decreased in the dextran transport and up-regulation of gene expression of tight junction molecules in human iPS-ECs. Moreover, Wnt inhibitors attenuated the effects of C6CM. In summary, we have established a simple protocol of the generation of BMEC-like cells from human iPSCs, and have demonstrated that differentiation of iPS-ECs to BMEC-like cells is induced by C6CM-derived signals, including canonical Wnt signals.


Assuntos
Neoplasias Encefálicas/patologia , Encéfalo/irrigação sanguínea , Glioma/patologia , Células-Tronco Pluripotentes Induzidas/citologia , Diferenciação Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Meios de Cultivo Condicionados , Meios de Cultura Livres de Soro , Humanos , Via de Sinalização Wnt
5.
Stem Cells Transl Med ; 4(5): 424-36, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25762001

RESUMO

In developing embryos or in vitro differentiation cultures using pluripotent stem cells (PSCs), such as embryonic stem cells and induced pluripotent stem cells, fetal liver kinase 1 (Flk1)-expressing mesodermal cells are thought to be a heterogeneous population that includes hematopoietic progenitors, endothelial progenitors, and cardiac progenitors. However, information on cell surface markers for separating these progenitors in Flk1⁺ cells is currently limited. In the present study, we show that distinct types of progenitor cells in Flk1⁺ cells could be separated according to the expression of coxsackievirus and adenovirus receptor (CAR, also known as CXADR), a tight junction component molecule. We found that mouse and human PSC- and mouse embryo-derived Flk1⁺ cells could be subdivided into Flk1⁺CAR⁺ cells and Flk1⁺CAR⁻ cells. The progenitor cells with cardiac potential were almost entirely restricted to Flk1⁺CAR⁺ cells, and Flk1⁺CAR⁻ cells efficiently differentiated into hematopoietic cells. Endothelial differentiation potential was observed in both populations. Furthermore, from the expression of CAR, Flk1, and platelet-derived growth factor receptor-α (PDGFRα), Flk1⁺ cells could be separated into three populations (Flk1⁺PDGFRα⁻ CAR⁻ cells, Flk1⁺PDGFRα⁻CAR⁺ cells, and Flk1⁺PDGFRα⁺CAR⁺ cells). Flk1⁺PDGFRα⁺ cells and Flk1⁺PDGFRα⁻ cells have been reported as cardiac and hematopoietic progenitor cells, respectively. We identified a novel population (Flk1⁺PDGFRα⁻ CAR⁺ cells) with the potential to differentiate into not only hematopoietic cells and endothelial cells but also cardiomyocytes. Our findings indicate that CAR would be a novel and prominent marker for separating PSC- and embryo-derived Flk1⁺ mesodermal cells with distinct differentiation potentials.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Hematopoéticas/metabolismo , Receptores Virais/biossíntese , Animais , Diferenciação Celular/genética , Linhagem da Célula , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/citologia , Regulação Viral da Expressão Gênica/genética , Humanos , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Receptores Virais/genética
6.
Cell Transplant ; 24(6): 1127-38, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24806294

RESUMO

Human liver chimeric mice are expected to be applied for drug toxicity tests and human hepatitis virus research. Human induced pluripotent stem cell-derived hepatocyte-like cells (iPSC-HLCs) are a highly attractive donor source for the generation of human liver chimeric mice because they can be produced on a large scale and established from an individual. Although these cells have been successfully used to generate human liver chimeric mice, there is still room for improvement in the repopulation efficiency. To enhance the repopulation efficacy, the human iPSC-HLCs were transduced with an adenovirus vector (Ad-FNK) expressing FNK, a hyperactive mutant gene from Bcl-xL, which was expected to inhibit apoptosis in the process of integration into liver parenchyma. We then transplanted Ad-FNK-transduced human iPSC-HLCs into urokinase-type plasminogen activator-transgenic severe combined immunodeficiency (uPA/SCID) mice (FNK mice) and evaluated the repopulation efficacy. The antiapoptotic effects of the human iPSC-HLCs were enhanced by FNK overexpression in vitro. Human albumin levels in the transplanted mice were significantly increased by transplantation of Ad-FNK-transduced human iPSC-HLCs (about 24,000 ng/ml). Immunohistochemical analysis with an anti-human αAT antibody revealed greater repopulation efficacy in the livers of FNK mice than control mice. Interestingly, the expression levels of human hepatocyte-related genes in the human iPSC-HLCs of FNK mice were much higher than those in the human iPSC-HLCs before transplantation. We succeeded in improving the repopulation efficacy of human liver chimeric mice generated by transplanting the Ad-FNK-transduced human iPSC-HLCs into uPA/SCID mice. Our method using ectopic expression of FNK was useful for generating human chimeric mice with high chimerism.


Assuntos
Hepatócitos/transplante , Células-Tronco Pluripotentes Induzidas/transplante , Mutação/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteína bcl-X/genética , Albuminas/metabolismo , Animais , Apoptose , Linhagem Celular , Hepatócitos/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Fígado/metabolismo , Masculino , Camundongos SCID , Reação em Cadeia da Polimerase em Tempo Real , Transdução Genética , Regulação para Cima
7.
PLoS One ; 9(3): e90791, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24651531

RESUMO

Human embryonic stem cells (hESCs) could provide a major window into human developmental biology, because the differentiation methods from hESCs mimic human embryogenesis. We previously reported that the overexpression of hematopoietically expressed homeobox (HHEX) in the hESC-derived definitive endoderm (DE) cells markedly promotes hepatic specification. However, it remains unclear how HHEX functions in this process. To reveal the molecular mechanisms of hepatic specification by HHEX, we tried to identify the genes directly targeted by HHEX. We found that HHEX knockdown considerably enhanced the expression level of eomesodermin (EOMES). In addition, HHEX bound to the HHEX response element located in the first intron of EOMES. Loss-of-function assays of EOMES showed that the gene expression levels of hepatoblast markers were significantly upregulated, suggesting that EOMES has a negative role in hepatic specification from the DE cells. Furthermore, EOMES exerts its effects downstream of HHEX in hepatic specification from the DE cells. In conclusion, the present results suggest that HHEX promotes hepatic specification by repressing EOMES expression.


Assuntos
Linhagem da Célula , Proteínas de Homeodomínio/metabolismo , Fígado/citologia , Proteínas com Domínio T/genética , Fatores de Transcrição/metabolismo , Animais , Biomarcadores/metabolismo , Padronização Corporal/efeitos dos fármacos , Padronização Corporal/genética , Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Endoderma/citologia , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células HeLa , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , RNA Interferente Pequeno/metabolismo , Elementos de Resposta/genética , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/genética , Transfecção , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
8.
Drug Metab Pharmacokinet ; 29(4): 296-304, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24492672

RESUMO

Cytochrome P450 3A4 (CYP3A4) plays a crucial role in the pharmacokinetic and safety profiles of drugs. However, it is difficult to properly predict the pharmacokinetics and hepatotoxicity of drugs in humans using data from experimental animals, because the catalytic activities of CYP3A4 and other drug-metabolizing enzymes differ between human and animal organs. In order to easily generate an animal model for proper evaluation of human CYP3A4-mediated drug metabolism, we developed a human CYP3A4-expressing adenovirus (Ad) vector based on our novel Ad vector exhibiting significantly lower hepatotoxicity (Ad-E4-122aT-hCYP3A4). Intravenous administration of Ad-E4-122aT-hCYP3A4 at a dose of 2 × 10(11) virus particles/mouse produced a mouse exhibiting human CYP3A4 activity at a level similar to that in the human liver, as shown in the dexamethasone metabolic experiment using liver microsomes. The area under the curve (AUC) of 6ßOHD was 2.7-fold higher in the Ad-E4-122aT-hCYP3A4-administered mice, compared with the mice receiving a control Ad vector. This Ad vector-expressing human CYP3A4 would thus be a powerful tool for evaluating human CYP3A4-mediated drug metabolism in the livers of experimental animals.


Assuntos
Adenovírus Humanos/genética , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Microssomos Hepáticos/metabolismo , Modelos Animais , Administração Intravenosa , Animais , Linhagem Celular , Dexametasona/metabolismo , Feminino , Humanos , Camundongos
9.
Development ; 141(1): 91-100, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24284203

RESUMO

Human embryonic stem cells (hESCs) and their derivatives are expected to be used in drug discovery, regenerative medicine and the study of human embryogenesis. Because hepatocyte differentiation from hESCs has the potential to recapitulate human liver development in vivo, we employed this differentiation method to investigate the molecular mechanisms underlying human hepatocyte differentiation. A previous study has shown that a gradient of transforming growth factor beta (TGFß) signaling is required to segregate hepatocyte and cholangiocyte lineages from hepatoblasts. Although CCAAT/enhancer binding proteins (c/EBPs) are known to be important transcription factors in liver development, the relationship between TGFß signaling and c/EBP-mediated transcriptional regulation in the hepatoblast fate decision is not well known. To clarify this relationship, we examined whether c/EBPs could determine the hepatoblast fate decision via regulation of TGFß receptor 2 (TGFBR2) expression in the hepatoblast-like cells differentiated from hESCs. We found that TGFBR2 promoter activity was negatively regulated by c/EBPα and positively regulated by c/EBPß. Moreover, c/EBPα overexpression could promote hepatocyte differentiation by suppressing TGFBR2 expression, whereas c/EBPß overexpression could promote cholangiocyte differentiation by enhancing TGFBR2 expression. Our findings demonstrated that c/EBPα and c/EBPß determine the lineage commitment of hepatoblasts by negatively and positively regulating the expression of a common target gene, TGFBR2, respectively.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Hepatócitos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/biossíntese , Proteína beta Intensificadora de Ligação a CCAAT/biossíntese , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Fígado/embriologia , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Camundongos , Regiões Promotoras Genéticas/genética , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo
10.
Stem Cells Dev ; 23(18): 2202-10, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24344904

RESUMO

Vascular endothelial growth factor (VEGF) is reported to exhibit potent hematopoietic stem/progenitor cell (HSPC) mobilization activity. However, the detailed mechanisms of HSPC mobilization by VEGF have not been examined. In this study, we investigated the effect of VEGF on bone marrow (BM) cell and the BM environment by intravenous injection of VEGF-expressing adenovirus vector (Ad-VEGF) into mice. A colony assay using peripheral blood cells revealed that plasma elevation of VEGF leads to the mobilization of HSPCs into the circulation. Granulocyte colony-stimulating factor (G-CSF) is known to mobilize HSPCs by decreasing CXC chemokine ligand 12 (CXCL12) levels in the BM. However, we found almost no changes in the CXCL12 levels in the BM after Ad-VEGF injection, suggesting that VEGF can alter the BM microenvironment by different mechanisms from G-CSF. Furthermore, flow cytometric analysis and colony forming unit-fibroblast assay showed a reduction in the number of mesenchymal progenitor cells (MPCs), which have been reported to serve as niche cells to support HSPCs, in the BM of Ad-VEGF-injected mice. Adhesion of donor cells to the recipient BM after transplantation was also impaired in mice injected with Ad-VEGF, suggesting a decrease in the niche cell number. We also observed a dose-dependent chemoattractive effect of VEGF on primary BM stromal cells in vitro. These data suggest that VEGF alters the distribution of MPCs in the BM and can also mobilize MPCs to peripheral tissues. Taken together, our results imply that VEGF-elicited egress of HSPCs would be mediated, in part, by changing the number of MPCs in the BM.


Assuntos
Medula Óssea/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Mesenquimais/citologia , Fator A de Crescimento do Endotélio Vascular/sangue , Adenoviridae/metabolismo , Animais , Contagem de Células , Movimento Celular , Quimiocina CXCL12/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos
11.
Stem Cell Reports ; 1(4): 322-35, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24319667

RESUMO

The establishment of self-renewing hepatoblast-like cells (HBCs) from human pluripotent stem cells (PSCs) would realize a stable supply of hepatocyte-like cells for medical applications. However, the functional characterization of human PSC-derived HBCs was not enough. To purify and expand human PSC-derived HBCs, human PSC-derived HBCs were cultured on dishes coated with various types of human recombinant laminins (LN). Human PSC-derived HBCs attached to human laminin-111 (LN111)-coated dish via integrin alpha 6 and beta 1 and were purified and expanded by culturing on the LN111-coated dish, but not by culturing on dishes coated with other laminin isoforms. By culturing on the LN111-coated dish, human PSC-derived HBCs were maintained for more than 3 months and had the ability to differentiate into both hepatocyte-like cells and cholangiocyte-like cells. These expandable human PSC-derived HBCs would be manageable tools for drug screening, experimental platforms to elucidate mechanisms of hepatoblasts, and cell sources for hepatic regenerative therapy.


Assuntos
Técnicas de Cultura de Células , Células-Tronco Pluripotentes/citologia , Animais , Adesão Celular , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Transplante de Células , Hepatócitos/citologia , Hepatócitos/transplante , Humanos , Integrina alfa6/metabolismo , Integrina beta1/metabolismo , Laminina/farmacologia , Camundongos
12.
Stem Cells Dev ; 22(5): 726-34, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23045993

RESUMO

Mast cells play important roles in the pathogenesis of allergic diseases. They are generally classified into 2 phenotypically distinct populations: connective tissue-type mast cells (CTMCs) and mucosal-type mast cells (MMCs). The number of mast cells that can be obtained from tissues is limited, making it difficult to study the function of mast cells. Here, we report the generation and characterization of CTMC-like mast cells derived from mouse induced pluripotent stem (iPS) cells. iPS cell-derived mast cells (iPSMCs) were generated by the OP9 coculture method or embryoid body formation method. The number of Safranin O-positive cells, expression levels of CD81 protein and histidine decarboxylase mRNA, and protease activities were elevated in the iPSMCs differentiated by both methods as compared with those in bone marrow-derived mast cells (BMMCs). Electron microscopic analysis revealed that iPSMCs contained more granules than BMMCs. Degranulation was induced in iPSMCs after stimulation with cationic secretagogues or vancomycin. In addition, iPSMCs had the ability to respond to stimulation with the IgE/antigen complex in vitro and in vivo. Moreover, when iPSMCs generated on OP9 cells were cocultured with Swiss 3T3 fibroblasts, protease activities as maturation index were more elevated, demonstrating that mature mast cells were differentiated from iPS cells. iPSMCs can be used as an in vitro model of CTMCs to investigate their functions.


Assuntos
Diferenciação Celular , Células do Tecido Conjuntivo/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Mastócitos/citologia , Células 3T3 , Animais , Degranulação Celular , Linhagem Celular , Histidina Descarboxilase/genética , Imunoglobulina E/imunologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Mastócitos/imunologia , Mastócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Anafilaxia Cutânea Passiva , Fenazinas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tetraspanina 28/genética , Vancomicina/farmacologia
13.
Biomaterials ; 34(7): 1781-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23228427

RESUMO

Although it is expected that hepatocyte-like cells differentiated from human embryonic stem (ES) cells or induced pluripotent stem (iPS) cells will be utilized in drug toxicity testing, the actual applicability of hepatocyte-like cells in this context has not been well examined so far. To generate mature hepatocyte-like cells that would be applicable for drug toxicity testing, we established a hepatocyte differentiation method that employs not only stage-specific transient overexpression of hepatocyte-related transcription factors but also a three-dimensional spheroid culture system using a Nanopillar Plate. We succeeded in establishing protocol that could generate more matured hepatocyte-like cells than our previous protocol. In addition, our hepatocyte-like cells could sensitively predict drug-induced hepatotoxicity, including reactive metabolite-mediated toxicity. In conclusion, our hepatocyte-like cells differentiated from human ES cells or iPS cells have potential to be applied in drug toxicity testing.


Assuntos
Células-Tronco Embrionárias/citologia , Hepatócitos/efeitos dos fármacos , Células-Tronco Pluripotentes/citologia , Testes de Toxicidade/métodos , Diferenciação Celular , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica
14.
Stem Cells Dev ; 21(18): 3381-90, 2012 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-22738147

RESUMO

Embryonic stem (ES) cell- and induced pluripotent stem (iPS) cell-derived hematopoietic stem/progenitor cells (HSPCs) are considered as an unlimited source for HSPC transplantation. However, production of immature hematopoietic cells, especially HSPCs, from ES and iPS cells has been challenging. The adaptor protein Lnk has been shown to negatively regulate HSPC function via the inhibition of thrombopoietin (TPO) and stem cell factor signaling, and Lnk-deficient HSPCs show an enhanced self-renewal and repopulation capacity. In this study, we examined the role of Lnk on the hematopoietic differentiation from mouse ES and iPS cells by the inhibition of Lnk using a dominant-negative mutant of the Lnk (DN-Lnk) gene. We generated mouse ES and iPS cells stably expressing a DN-Lnk, and found that enforced expression of a DN-Lnk in ES and iPS cells led to an enhanced generation of Flk-1-positive mesodermal cells, thereby could increase in the expression of hematopoietic transcription factors, including Scl and Runx1. We also showed that the number of both total hematopoietic cells and immature hematopoietic cells with colony-forming potential in DN-Lnk-expressing cells was significantly increased in comparison with that in control cells. Furthermore, Lnk inhibition by the overexpression of the DN-Lnk gene augmented the TPO-induced phosphorylation of Erk1/2 and Akt, indicating the enhanced sensitivity to TPO. Adenovirus vector-mediated transient DN-Lnk gene expression in ES and iPS cells could also increase the hematopoietic cell production. Our data clearly showed that the inhibition of Lnk in ES and iPS cells could result in the efficient generation and expansion of hematopoietic cells.


Assuntos
Diferenciação Celular/genética , Células-Tronco Hematopoéticas , Células-Tronco Pluripotentes Induzidas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Linhagem Celular , Subunidade alfa 2 de Fator de Ligação ao Core/biossíntese , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas de Membrana , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Trombopoetina/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese
15.
J Hepatol ; 57(3): 628-36, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22659344

RESUMO

BACKGROUND & AIMS: Hepatocyte-like cells differentiated from human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) can be utilized as a tool for screening for hepatotoxicity in the early phase of pharmaceutical development. We have recently reported that hepatic differentiation is promoted by sequential transduction of SOX17, HEX, and HNF4α into hESC- or hiPSC-derived cells, but further maturation of hepatocyte-like cells is required for widespread use of drug screening. METHODS: To screen for hepatic differentiation-promoting factors, we tested the seven candidate genes related to liver development. RESULTS: The combination of two transcription factors, FOXA2 and HNF1α, promoted efficient hepatic differentiation from hESCs and hiPSCs. The expression profile of hepatocyte-related genes (such as genes encoding cytochrome P450 enzymes, conjugating enzymes, hepatic transporters, and hepatic nuclear receptors) achieved with FOXA2 and HNF1α transduction was comparable to that obtained in primary human hepatocytes. The hepatocyte-like cells generated by FOXA2 and HNF1α transduction exerted various hepatocyte functions including albumin and urea secretion, and the uptake of indocyanine green and low density lipoprotein. Moreover, these cells had the capacity to metabolize all nine tested drugs and were successfully employed to evaluate drug-induced cytotoxicity. CONCLUSIONS: Our method employing the transduction of FOXA2 and HNF1α represents a useful tool for the efficient generation of metabolically functional hepatocytes from hESCs and hiPSCs, and the screening of drug-induced cytotoxicity.


Assuntos
Células-Tronco Embrionárias/citologia , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/genética , Hepatócitos/citologia , Hepatócitos/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Bupropiona/metabolismo , Diferenciação Celular , Linhagem Celular , Células-Tronco Embrionárias/metabolismo , Etanolaminas/metabolismo , Técnicas de Transferência de Genes , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Hepatócitos/enzimologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Midazolam/metabolismo , Paclitaxel/metabolismo , Fenacetina/metabolismo , Testosterona/metabolismo , Tolbutamida/metabolismo , Transdução Genética
16.
Biomaterials ; 33(18): 4526-34, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22445253

RESUMO

Hepatocyte-like cells differentiated from human embryonic stem cells (hESCs) or human induced pluripotent stem cells (hiPSCs) are known to be a useful cell source for drug screening. We recently developed an efficient hepatic differentiation method from hESCs and hiPSCs by sequential transduction of FOXA2 and HNF1α. It is known that the combination of three-dimensional (3D) culture and co-culture, namely 3D co-culture, can maintain the functions of primary hepatocytes. However, hepatic maturation of hESC- or hiPSC-derived hepatocyte-like cells (hEHs or hiPHs, respectively) by 3D co-culture systems has not been examined. Therefore, we utilized a cell sheet engineering technology to promote hepatic maturation. The gene expression levels of hepatocyte-related markers (such as cytochrome P450 enzymes and conjugating enzymes) and the amount of albumin secretion in the hEHs or hiPHs, which were 3D co-cultured with the Swiss 3T3 cell sheet, were significantly up-regulated in comparison with those in the hEHs or hiPHs cultured in a monolayer. Furthermore, we found that type I collagen synthesized in Swiss 3T3 cells plays an important role in hepatic maturation. The hEHs or hiPHs that were 3D co-cultured with the Swiss 3T3 cell sheet would be powerful tools for medical applications, such as drug screening.


Assuntos
Técnicas de Cocultura/métodos , Colágeno Tipo I/metabolismo , Fígado/citologia , Células-Tronco Pluripotentes/citologia , Animais , Técnicas de Cultura de Células/métodos , Linhagem Celular , Hepatócitos/citologia , Humanos , Camundongos , Células Swiss 3T3
17.
Stem Cell Res ; 8(2): 300-11, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22000550

RESUMO

Ectopic expression of HoxB4 in embryonic stem (ES) cells leads to an efficient production of hematopoietic cells, including hematopoietic stem/progenitor cells. Previous studies have utilized a constitutive HoxB4 expression system or tetracycline-regulated HoxB4 expression system to induce hematopoietic cells from ES cells. However, these methods cannot be applied therapeutically due to the risk of transgenes being integrated into the host genome. Here, we report the promotion of hematopoietic differentiation from mouse ES cells and induced pluripotent stem (iPS) cells by transient HoxB4 expression using an adenovirus (Ad) vector. Ad vector could mediate efficient HoxB4 expression in ES cell-derived embryoid bodies (ES-EBs) and iPS-EBs, and its expression was decreased during cultivation, showing that Ad vector transduction was transient. A colony-forming assay revealed that the number of hematopoietic progenitor cells with colony-forming potential in HoxB4-transduced cells was significantly increased in comparison with that in non-transduced cells or LacZ-transduced cells. HoxB4-transduced cells also showed more efficient generation of CD41-, CD45-, or Sca-1-positive cells than control cells. These results indicate that transient, but not constitutive, HoxB4 expression is sufficient to augment the hematopoietic differentiation of ES and iPS cells, and that our method would be useful for clinical applications, such as cell transplantation therapy.


Assuntos
Diferenciação Celular , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Fatores de Transcrição/metabolismo , Transdução Genética , Adenoviridae/genética , Animais , Antígenos de Superfície/metabolismo , Biomarcadores/metabolismo , Contagem de Células , Ensaio de Unidades Formadoras de Colônias , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos
18.
Mol Ther ; 20(1): 127-37, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22068426

RESUMO

Hepatocyte-like cells from human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) are expected to be a useful source of cells drug discovery. Although we recently reported that hepatic commitment is promoted by transduction of SOX17 and HEX into human ESC- and iPSC-derived cells, these hepatocyte-like cells were not sufficiently mature for drug screening. To promote hepatic maturation, we utilized transduction of the hepatocyte nuclear factor 4α (HNF4α) gene, which is known as a master regulator of liver-specific gene expression. Adenovirus vector-mediated overexpression of HNF4α in hepatoblasts induced by SOX17 and HEX transduction led to upregulation of epithelial and mature hepatic markers such as cytochrome P450 (CYP) enzymes, and promoted hepatic maturation by activating the mesenchymal-to-epithelial transition (MET). Thus HNF4α might play an important role in the hepatic differentiation from human ESC-derived hepatoblasts by activating the MET. Furthermore, the hepatocyte like-cells could catalyze the toxication of several compounds. Our method would be a valuable tool for the efficient generation of functional hepatocytes derived from human ESCs and iPSCs, and the hepatocyte-like cells could be used for predicting drug toxicity.


Assuntos
Células-Tronco Embrionárias/citologia , Fator 4 Nuclear de Hepatócito/genética , Hepatócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Transdução Genética , Diferenciação Celular , Linhagem Celular , Células-Tronco Embrionárias/metabolismo , Transição Epitelial-Mesenquimal/genética , Técnicas de Transferência de Genes , Fator 4 Nuclear de Hepatócito/metabolismo , Hepatócitos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Fatores de Tempo
19.
Yakugaku Zasshi ; 131(9): 1333-8, 2011.
Artigo em Japonês | MEDLINE | ID: mdl-21881308

RESUMO

Because embryonic stem (ES) cells and induced pluripotent stem (iPS) cells can differentiate into various types of cells in vitro, they are considered as a valuable model to understand the processes involved in the differentiation into functional cells as well as an unlimited source of cells for therapeutic applications. Efficient gene transduction method is one of the powerful tools for the basic researches and for differentiating ES and iPS cells into lineage-committed cells. Recently, we have developed an adenovirus (Ad) vector for efficient transduction into ES and iPS cells. We showed that Ad vectors containing the cytomegalovirus enhancer/ß-actin promoter with ß-actin intron (CA) promoter or the elongation factor (EF)-1α promoter were the appropriate for the transduction into ES and iPS cells. We also found that enforced expression of a PPARγ gene or a Runx2 gene into mouse ES and iPS cells by an optimized Ad vector markedly augmented the differentiation of adipocytes or osteoblasts, respectively. Thus, a gene transfer technique using an Ad vector could be an advantage for the regulation of stem cell differentiation and could be applied to regenerative medicine based on ES and iPS cells.


Assuntos
Adenoviridae , Diferenciação Celular/genética , Células-Tronco Embrionárias/citologia , Técnicas de Transferência de Genes , Vetores Genéticos , Células-Tronco Pluripotentes Induzidas/citologia , Adenoviridae/química , Animais , Vetores Genéticos/química , Humanos , Subunidades beta de Inibinas , Camundongos , Fator 1 de Elongação de Peptídeos , Regiões Promotoras Genéticas , Medicina Regenerativa/métodos
20.
PLoS One ; 6(7): e21780, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21760905

RESUMO

The establishment of methods for directive differentiation from human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) is important for regenerative medicine. Although Sry-related HMG box 17 (SOX17) overexpression in ESCs leads to differentiation of either extraembryonic or definitive endoderm cells, respectively, the mechanism of these distinct results remains unknown. Therefore, we utilized a transient adenovirus vector-mediated overexpression system to mimic the SOX17 expression pattern of embryogenesis. The number of alpha-fetoprotein-positive extraembryonic endoderm (ExEn) cells was increased by transient SOX17 transduction in human ESC- and iPSC-derived primitive endoderm cells. In contrast, the number of hematopoietically expressed homeobox (HEX)-positive definitive endoderm (DE) cells, which correspond to the anterior DE in vivo, was increased by transient adenovirus vector-mediated SOX17 expression in human ESC- and iPSC-derived mesendoderm cells. Moreover, hepatocyte-like cells were efficiently generated by sequential transduction of SOX17 and HEX. Our findings show that a stage-specific transduction of SOX17 in the primitive endoderm or mesendoderm promotes directive ExEn or DE differentiation by SOX17 transduction, respectively.


Assuntos
Diferenciação Celular , Linhagem da Célula , Células-Tronco Embrionárias/citologia , Endoderma/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Fatores de Transcrição SOXF/metabolismo , Transdução Genética , Adenoviridae/metabolismo , Linhagem Celular , Células-Tronco Embrionárias/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fígado/citologia , Mesoderma/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...