Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
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.
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
3.
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
4.
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
5.
Mol Ther ; 19(2): 400-7, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21102561

RESUMO

Human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have the potential to differentiate into all cell lineages, including hepatocytes, in vitro. Induced hepatocytes have a wide range of potential application in biomedical research, drug discovery, and the treatment of liver disease. However, the existing protocols for hepatic differentiation of PSCs are not very efficient. In this study, we developed an efficient method to induce hepatoblasts, which are progenitors of hepatocytes, from human ESCs and iPSCs by overexpression of the HEX gene, which is a homeotic gene and also essential for hepatic differentiation, using a HEX-expressing adenovirus (Ad) vector under serum/feeder cell-free chemically defined conditions. Ad-HEX-transduced cells expressed α-fetoprotein (AFP) at day 9 and then expressed albumin (ALB) at day 12. Furthermore, the Ad-HEX-transduced cells derived from human iPSCs also produced several cytochrome P450 (CYP) isozymes, and these P450 isozymes were capable of converting the substrates to metabolites and responding to the chemical stimulation. Our differentiation protocol using Ad vector-mediated transient HEX transduction under chemically defined conditions efficiently generates hepatoblasts from human ESCs and iPSCs. Thus, our methods would be useful for not only drug screening but also therapeutic applications.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Genes Homeobox/fisiologia , Hepatócitos/citologia , Proteínas de Homeodomínio/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Fatores de Transcrição/fisiologia , Adenoviridae/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Genes Homeobox/genética , Vetores Genéticos/genética , Proteínas de Homeodomínio/genética , Humanos , Fatores de Transcrição/genética
6.
Stem Cells ; 27(8): 1802-11, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19544436

RESUMO

Induced pluripotent stem (iPS) cells, which are generated from somatic cells by transducing four genes, are expected to have broad application to regenerative medicine. Although establishment of an efficient gene transfer system for iPS cells is considered to be essential for differentiating them into functional cells, the detailed transduction characteristics of iPS cells have not been examined. Previously, by using an adenovirus (Ad) vector containing the elongation factor-1alpha (EF-1alpha) and the cytomegalovirus enhancer/beta-actin (CA) promoters, we developed an efficient transduction system for mouse embryonic stem (ES) cells and their aggregate form, embryoid bodies (EBs). In this study, we applied our transduction system to mouse iPS cells and investigated whether efficient differentiation could be achieved by Ad vector-mediated transduction of a functional gene. As in the case of ES cells, the Ad vector containing EF-1alpha and the CA promoter could efficiently transduce transgenes into mouse iPS cells. At 3,000 vector particles/cell, 80%-90% of iPS cells expressed transgenes by treatment with an Ad vector containing the CA promoter, without a decrease in pluripotency or viability. We also found that the CA promoter had potent transduction ability in iPS cell-derived EBs. Moreover, exogenous expression of a PPARgamma gene or a Runx2 gene into mouse iPS cells by an optimized Ad vector enhanced adipocyte or osteoblast differentiation, respectively. These results suggest that Ad vector-mediated transient transduction is sufficient to increase cellular differentiation and that our transduction methods would be useful for therapeutic applications based on iPS cells.


Assuntos
Adipócitos/citologia , Técnicas de Transferência de Genes , Osteoblastos/citologia , Células-Tronco Pluripotentes/citologia , Transdução Genética/métodos , Adenoviridae/genética , Adipócitos/metabolismo , Animais , Diferenciação Celular/fisiologia , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Expressão Gênica , Vetores Genéticos/genética , Camundongos , Microscopia Confocal , Osteoblastos/metabolismo , PPAR gama/genética , Fator 1 de Elongação de Peptídeos/genética , Células-Tronco Pluripotentes/metabolismo , Regiões Promotoras Genéticas , Receptores Virais/biossíntese , Receptores Virais/genética , Transgenes
7.
Yakugaku Zasshi ; 130(11): 1527-34, 2010 Nov.
Artigo em Japonês | MEDLINE | ID: mdl-21048413

RESUMO

Induced pluripotent stem (iPS) cells, which are generated from somatic cells by transducing four genes, are expected to have broad application to regenerative medicine. Although establishment of an efficient gene transfer system for iPS cells is considered to be essential for differentiating them into functional cells, the detailed transduction characteristics of iPS cells have not been examined. By using an adenovirus (Ad) vector containing the cytomegalovirus enhancer/beta-actin (CA) promoters, we have developed an efficient transduction system for mouse mesenchymal stem cells and embryonic stem (ES) cells. Also, we applied our transduction system to mouse iPS cells and investigated whether efficient differentiation could be achieved by Ad vector-mediated transduction of a functional gene. As in the case of ES cells, the Ad vector could efficiently transduce transgenes into mouse iPS cells. We found that the CA promoter had potent transduction ability in iPS cells. Moreover, exogenous expression of a PPARγ gene or a Runx2 gene into mouse iPS cells by an optimized Ad vector enhanced adipocyte or osteoblast differentiation, respectively. These results suggest that Ad vector-mediated transient transduction is sufficient to promote cellular differentiation and that our transduction methods would be useful for therapeutic applications based on iPS cells.


Assuntos
Diferenciação Celular , Técnicas de Transferência de Genes , Células-Tronco Pluripotentes Induzidas/citologia , Actinas , Adenoviridae , Adipócitos , Animais , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Células-Tronco Embrionárias , Vetores Genéticos , Células-Tronco Mesenquimais , Camundongos , Osteoblastos , PPAR gama/genética , Transdução Genética/métodos , Transgenes
8.
Biochem Biophys Res Commun ; 379(1): 127-32, 2009 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-19103162

RESUMO

Bone marrow stromal cells (BMSCs) are expected to be a source for tissue regeneration because they can differentiate into multiple cell types. Establishment of efficient gene transfer systems for BMSCs is essential for their application to regenerative medicine. In this study, we compared the transduction efficiency in mouse primary BMSCs by using fiber-modified adenovirus (Ad) vectors, and demonstrated that AdK7, which harbors a polylysin (K7) peptide in the C-terminus of the fiber knob, could efficiently express a transgene in BMSCs. Notably, AdK7 robustly drove transgene expression in more than 90% of the BMSCs at 3,000 vector particles/cell. Furthermore, we showed that in vitro and in vivo osteogenic potential of BMSCs was dramatically promoted by the transduction of Runx2 gene using AdK7. These results indicate that this transduction system could be a powerful tool for therapeutic applications based on BMSCs.


Assuntos
Células da Medula Óssea/citologia , Técnicas de Cultura de Células , Diferenciação Celular , Osteoblastos/fisiologia , Transdução Genética/métodos , Adenoviridae/genética , Animais , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Vetores Genéticos/química , Vetores Genéticos/genética , Camundongos , Osteoblastos/citologia , Osteogênese , Polilisina/química , Células Estromais/citologia
9.
J Gene Med ; 10(5): 498-507, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18278825

RESUMO

BACKGROUND: Establishment of a transient gene delivery system, such as adenovirus (Ad) vectors, into embryonic stem (ES) cells and their aggregation form, embryoid bodies (EBs), is essential for its application in regenerative medicine because the transgene should not be integrated in the host genome. In this study, we optimized Ad vector-mediated transduction into EBs, and examined whether Ad vector-mediated transduction of adipogenesis-related gene into EBs could promote the adipocyte differentiation. METHODS: We prepared beta-galactosidase-expressing Ad vectors under the control of four different promoters (cytomegalovirus (CMV), rouse sarcoma virus, human elongation factor-1alpha, and CMV enhancer/beta-actin promoter (CA)) to estimate the transduction efficiency. Adipocyte differentiation efficiency by transduction of the PPAR gamma or C/EBP alpha gene into EBs was examined. RESULTS: Of the four promoters tested, the CA promoter exhibited the highest transduction efficiency in the EBs. However, Ad vector-mediated transduction was observed only in the periphery of the EBs. When repeated transduction by Ad vector was performed, gene expression was observed even in the interior of EBs as well. When EB-derived single cells were transduced by an Ad vector containing the CA promoter, more than 90% of the cells were transduced. Furthermore, Ad vector-mediated PPAR gamma gene transduction into EBs led to more efficient differentiation into adipocytes than could untransduced EBs, examined in terms of lipogenic enzyme activities and accumulation of the lipid droplets. CONCLUSIONS: Ad vector-mediated transduction into EBs could be a valuable tool for molecular switching of cell differentiation and could be applied to regenerative medicine.


Assuntos
Adipócitos/citologia , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , PPAR gama/genética , Transdução Genética/métodos , Adenoviridae/genética , Animais , Vetores Genéticos , Camundongos , PPAR gama/fisiologia , Medicina Regenerativa/métodos
10.
J Control Release ; 117(3): 430-7, 2007 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-17239467

RESUMO

Vectors for gene expression are the essential tools for both gene therapy and basic research. There are two groups of gene therapy vectors, viral and non-viral vectors. At present, toxicity triggered by vectors is one of the major concerns for clinical trials. In general, non-viral vectors, such as plasmid DNA-cationic liposome complex (lipoplex), are thought to be safer than viral vectors, such as adenovirus (Ad) vector, although lipoplex is less efficient in term of gene expression than the Ad vector. However, there has been no study directly comparing the gene expression efficiency and safety of viral and non-viral vectors. Here, we present evidence that the Ad vector shows much more efficient gene expression and is safer than lipoplex, at least with respect to the innate immune response. After being systemically administered to mice, the Ad vector showed a transduction efficiency that was 2 to 5 log orders higher than that of lipoplex, depending on the organ. On the other hand, surprisingly, the administration of lipoplex produced a greater amount of inflammatory cytokines such as interleukin-6, interleukin-12, and tumor necrosis factor-alpha than did the administration of the Ad vector, whereas a comparable level of hepatotoxicity was induced by these vectors. The production of inflammatory cytokines induced by the injection of lipoplex was reduced when the CpG motifs were removed completely from plasmid DNA. Thus, care should be taken to ensure the innate immune response induced by gene therapy vectors, especially lipoplex.


Assuntos
Adenoviridae/genética , Expressão Gênica/fisiologia , Vetores Genéticos/administração & dosagem , Imunidade Inata/fisiologia , Animais , Doença Hepática Induzida por Substâncias e Drogas/patologia , Citocinas/biossíntese , Portadores de Fármacos , Excipientes , Ácidos Graxos Monoinsaturados , Feminino , Vetores Genéticos/efeitos adversos , Interleucina-12/biossíntese , Interleucina-6/biossíntese , Lipossomos , Fígado/metabolismo , Fígado/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Inclusão em Parafina , Plasmídeos/química , Plasmídeos/genética , Compostos de Amônio Quaternário , Transdução Genética , Fator de Necrose Tumoral alfa/biossíntese
11.
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
12.
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
13.
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
14.
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
15.
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
16.
Mol Cell Endocrinol ; 226(1-2): 27-32, 2004 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-15489002

RESUMO

Transcription of the CYP24 (25-hydroxyvitamin D(3)-24-hydroxylase) gene is known to be induced by 1alpha,25-dihydroxyvitamin D3 (1,25(OH)2D3). We studied the induction kinetics in detail in human skin-derived fibroblasts. While the basal transcription of this gene was very low, addition of 1,25(OH)2D3 increased the mRNA level by 50-fold within 1h. The induction reached as high as 20000-fold after 12h. DNA microarray analysis also indicated that the induction ratio of the CYP24 gene is exceptionally high among 3800 human genes examined. The increase of mRNA was caused by stimulation of the transcription, but not by stabilization of mRNA. 24(R),25-dihydroxyvitamin D3 (24,25(OH)2D3), a compound metabolically related to 1,25(OH)2D3, also stimulated the CYP24 gene transcription, though at much higher concentrations. However, this stimulation was significantly augmented by synergistic actions of 24,25(OH)2D3 and 1,25(OH)2D3, suggesting that 24,25(OH)2D3 or its metabolites might be playing some roles in the regulation of CYP24 gene transcription.


Assuntos
Sistema Enzimático do Citocromo P-450/genética , Fibroblastos/enzimologia , Pele/enzimologia , Esteroide Hidroxilases/genética , Transcrição Gênica/efeitos dos fármacos , Vitamina D/metabolismo , 24,25-Di-Hidroxivitamina D 3/farmacologia , Calcitriol/farmacologia , Agonistas dos Canais de Cálcio/farmacologia , Células Cultivadas , Sistema Enzimático do Citocromo P-450/metabolismo , Sinergismo Farmacológico , Ativação Enzimática/efeitos dos fármacos , Perfilação da Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Esteroide Hidroxilases/metabolismo , Vitamina D3 24-Hidroxilase
17.
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
18.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa