Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Biosci Bioeng ; 129(3): 371-378, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31615734

ABSTRACT

We optimized the conditions for the differentiation of human induced pluripotent stem cells (hiPSCs) into mesoderm lineage-committed cells by supplementing the cultures with CHIR, a selective GSK-3 inhibitor, during embryoid body (EB) formation. In vitro treatment with 4 µM CHIR during the late 2 days of a 4-day suspension culture period was most effective at promoting mesodermal differentiation. The resulting EBs showed a significant increase in the expression levels of mesoderm-associated genes (WNT3A, T, DKK1, GATA4, FOXC1, and MESP1) and a maintenance of OCT3/4 and NANOG expressions. Upon subsequent differentiation into a cardiac cell lineage, these EBs were shown to generate contractile cardiomyocytes. When shortening the CHIR treatment period to 1 day, the resulting EBs showed reduced expression of mesoderm-associated genes in comparison to the 2-day CHIR treatment. In particular, the expression level of FOXC1 in the 1-day CHIR-treated EBs was much lower than that of the 2-day CHIR-treated EBs. When the treatment period with CHIR was extended to 4 days, the resulting EBs presented significantly reduced expression of WNT3A, OCT3/4, and NANOG upon CHIR concentrations above 4 µM. Similarly, when CHIR treatment was conducted after the formation of EBs, the effectiveness of the GSK-3 inhibitor was reduced compared to a treatment performed during EB formation. Our results indicate that spatiotemporal constraints associated with EB formation, i.e., three-dimensional structuration and cell development in EBs, should be taken into account when designing EB formation-based differentiation protocol involving CHIR treatment.


Subject(s)
Cell Differentiation/drug effects , Embryoid Bodies/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Induced Pluripotent Stem Cells/drug effects , Mesoderm/drug effects , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Cell Line , Cell Lineage/drug effects , Embryoid Bodies/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Mesoderm/cytology , Myocytes, Cardiac/cytology
2.
Cornea ; 38 Suppl 1: S34-S41, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31403532

ABSTRACT

In its early stages, an embryo polarizes to form cell subpopulations that subsequently produce specific organ cell types. These cell subpopulations are defined by transcription factors (TFs) that activate or repress specific genes. Although an embryo comprises thousands of TFs, surprisingly few are needed to determine the fate of a given cell. The ectoderm divides into the neuroectoderm and surface ectoderm, the latter of which gives rise to epidermal keratinocytes and corneal epithelial cells (CECs). Meanwhile, neuroectoderm cells give rise to other parts of the eye such as the corneal endothelium and retina. To investigate the regulatory role of TFs in CECs, we overexpressed the "core TFs" (PAX6, OVOL2, and KLF4) in human fibroblasts and found that the cells adopted a CEC-like quality. OVOL2 overexpression was even able to directly induce cells with a neuroectoderm fate toward a surface ectoderm fate, designated "direct reprogramming." Conversely, suppression of OVOL2 or PAX6 expression induced CECs to show qualities consistent with neural lineage cells or epidermal keratinocytes, respectively. This suggests that these core TFs can maintain the CEC phenotype through reciprocal gene regulation. Direct reprogramming has important implications for cell therapies. The potential benefits of cells derived by direct reprogramming compared with induced pluripotent stem cells include the fact that it requires less time than reprogramming a cell back to the pluripotent state and then to another cell type. Further understanding of the reciprocally repressive mechanism of action for core TFs could lead to alternative treatments for regenerative medicine not requiring cell transplantation.


Subject(s)
Epithelium, Corneal/metabolism , Gene Expression Regulation , Kruppel-Like Transcription Factors/genetics , PAX6 Transcription Factor/genetics , Transcription Factors/genetics , Animals , Cell Differentiation , Cell Lineage , Epithelium, Corneal/cytology , Gene Regulatory Networks , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/biosynthesis , PAX6 Transcription Factor/biosynthesis , Transcription Factors/biosynthesis
3.
Sci Rep ; 8(1): 8716, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29880849

ABSTRACT

Induced pluripotent stem cell (iPS) reprogramming allows to turn a differentiated somatic cell into a pluripotent cell. This process is accompanied by many changes in fundamental cell properties, such as energy production, cell-to-cell interactions, cytoskeletal organization, and others. Real-time quantitative polymerase chain reaction (RT-qPCR) can be used as a quantitative method of gene expression analysis to investigate iPS reprogramming but it requires a validation of reference genes for the accurate assessment of target genes' expression. Currently, studies evaluating the performance of reference genes during iPS reprogramming are lacking. In this study we analysed the stability of 12 housekeeping genes during 20 days of iPS reprogramming of murine cells based on statistical analyses of RT-qPCR data using five different statistical algorithms. This study reports strong variations in housekeeping gene stability during the reprogramming process. Most stable genes were Atp5f1, Pgk1 and Gapdh, while the least stable genes were Rps18, Hprt, Tbp and Actb. The results were validated by a proof-of-point qPCR experiment with pluripotent markers Nanog, Rex1 and Oct4 normalized to the best and the worst reference gene identified by the analyses. Overall, this study and its implications are particularly relevant to investigations on the cell-state and pluripotency in iPS reprogramming.


Subject(s)
Cellular Reprogramming , Gene Expression Profiling/methods , Gene Expression Regulation , Genes, Essential , Induced Pluripotent Stem Cells/metabolism , Real-Time Polymerase Chain Reaction/methods , Animals , Cellular Reprogramming Techniques , Gene Expression Profiling/standards , Humans , Mice , Real-Time Polymerase Chain Reaction/standards , Reference Standards
4.
Nat Commun ; 9(1): 1387, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29643333

ABSTRACT

Multicellular organisms consist of multiple cell types. The identity of these cells is primarily maintained by cell-type-specific gene expression programs; however, mechanisms that suppress these programs are poorly defined. Here we show that serum response factor (Srf), a transcription factor that is activated by various extracellular stimuli, can repress cell-type-specific genes and promote cellular reprogramming to pluripotency. Manipulations that decrease ß-actin monomer quantity result in the nuclear accumulation of Mkl1 and the activation of Srf, which downregulate cell-type-specific genes and alter the epigenetics of regulatory regions and chromatin organization. Mice overexpressing Srf exhibit various pathologies including an ulcerative colitis-like symptom and a metaplasia-like phenotype in the pancreas. Our results demonstrate an unexpected function of Srf via a mechanism by which extracellular stimuli actively destabilize cell identity and suggest Srf involvement in a wide range of diseases.


Subject(s)
Chromatin/chemistry , Colitis, Ulcerative/genetics , Induced Pluripotent Stem Cells/metabolism , Metaplasia/genetics , Neural Stem Cells/metabolism , Pancreas/metabolism , Serum Response Factor/genetics , Actins/genetics , Actins/metabolism , Animals , Cell Differentiation , Cellular Reprogramming/genetics , Chromatin/metabolism , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Induced Pluripotent Stem Cells/cytology , Male , Metaplasia/metabolism , Metaplasia/pathology , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Pancreas/pathology , Serum Response Factor/metabolism , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism
5.
Genes Cells ; 22(10): 918-928, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28776863

ABSTRACT

The molecular mechanisms of cell reprogramming and differentiation involve various signaling factors. Small molecule compounds have been identified to artificially influence these factors through interacting cellular proteins. Although such small molecule compounds are useful to enhance reprogramming and differentiation and to show the mechanisms that underlie these events, the screening usually requires a large number of compounds to identify only a very small number of hits (e.g., one hit among several tens of thousands of compounds). Here, we show a proof of concept that xenospecific gene products can affect the efficiency of cell reprogramming to pluripotency. Thirty genes specific for the bacterium Wolbachia pipientis were forcibly expressed individually along with reprogramming factors (Oct4, Sox2, Klf4 and c-Myc) that can generate induced pluripotent stem cells in mammalian cells, and eight were found to affect the reprogramming efficiency either positively or negatively (hit rate 26.7%). Mechanistic analysis suggested one of these proteins interacted with cytoskeleton to promote reprogramming. Our results raise the possibility that xenospecific gene products provide an alternative way to study the regulatory mechanism of cell identity.


Subject(s)
Cellular Reprogramming/genetics , Genes, Bacterial , Neural Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Line , Cytoskeleton/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Neural Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Wolbachia/genetics
6.
Exp Eye Res ; 154: 30-38, 2017 01.
Article in English | MEDLINE | ID: mdl-27818314

ABSTRACT

PAX6, a paired box transcription factor, is necessary for eye development. However, how it regulates the cell identity of human corneal epithelial cells (CECs) is not well understood. We aimed to clarify the function of PAX6 in human CECs using gene knockout via the clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR associated protein 9 (Cas9) system. We designed guide RNAs for different targets in PAX6. PAX6-depleted CECs maintained the epithelial morphology, but became larger. Global analyses using microarray revealed that down-regulated genes were primarily CEC-specific and included keratin 12, keratin 3, clusterin (CLU), aldehyde dehydrogenase 3 family member A1 (ALDH3A1), angiopoietin-like 7 (ANGPTL7) and transketolase (TKT), while up-regulated genes were primarily epidermis-related and included keratin 10, keratin 1, involucrin (IVL), filaggrin (FLG). These findings suggest that PAX6 maintains CEC identity by regulating differentiation.


Subject(s)
Epithelium, Corneal/metabolism , Gene Expression Regulation , PAX6 Transcription Factor/genetics , RNA/genetics , Blotting, Western , Cell Differentiation , Epithelium, Corneal/cytology , Filaggrin Proteins , Gene Knockout Techniques , Humans , Immunohistochemistry , Microarray Analysis , PAX6 Transcription Factor/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
7.
Cell Rep ; 15(6): 1359-68, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27134177

ABSTRACT

In development, embryonic ectoderm differentiates into neuroectoderm and surface ectoderm using poorly understood mechanisms. Here, we show that the transcription factor OVOL2 maintains the transcriptional program of human corneal epithelium cells (CECs), a derivative of the surface ectoderm, and that OVOL2 may regulate the differential transcriptional programs of the two lineages. A functional screen identified OVOL2 as a repressor of mesenchymal genes to maintain CECs. Transduction of OVOL2 with several other transcription factors induced the transcriptional program of CECs in fibroblasts. Moreover, neuroectoderm derivatives were found to express mesenchymal genes, and OVOL2 alone could induce the transcriptional program of CECs in neural progenitors by repressing these genes while activating epithelial genes. Our data suggest that the difference between the transcriptional programs of some neuroectoderm- and surface ectoderm-derivative cells may be regulated in part by a reciprocally repressive mechanism between epithelial and mesenchymal genes, as seen in epithelial-to-mesenchymal transition.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Epithelium, Corneal/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Epithelial Cells/metabolism , Epithelium, Corneal/cytology , Epithelium, Corneal/growth & development , Fibroblasts/metabolism , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/metabolism , Mesoderm/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism
8.
Biotechnol Lett ; 38(4): 731-40, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26721236

ABSTRACT

OBJECTIVES: To examine whether an autonomously replicating, artificial chromosome-like vector containing a long genomic DNA sequence (namely, Epigenosome-Nanog) undergoes de novo CpG methylation after maintenance in cultured cells for more than a half year. RESULTS: Epigenosome-Nanog efficiently replicated in iPS cells after transfection. In HeLa and C2C12 cells Epigenosome-Nanog was stably maintained for more than eight months. The CpG methylation occurred de novo at the Nanog gene promoter region on the epigenosome in C2C12 cells but the degrees of methylation were much lower than those at the same CpG sites on the chromosomes. Among the four CpG sites at the region, the upstream two CpGs underwent methylation in a correlated manner while methylation at the downstream two CpGs was also correlated to each other, and these correlations were commonly shared between the epigenosome and the chromosome. CpG methylation thus was not solely dependent on the nucleotide sequence at the DNA locus. CONCLUSION: The epigenosome may become a useful tool to study the mechanisms of epigenetic regulation of a genetic region of interest in mammalian cells.


Subject(s)
Chromosomes, Artificial/genetics , CpG Islands , DNA Methylation , Animals , Cell Culture Techniques , Cell Line , Epigenesis, Genetic , HeLa Cells , Humans , Mice , Promoter Regions, Genetic
9.
Nihon Rinsho ; 73 Suppl 5: 337-41, 2015 Jun.
Article in Japanese | MEDLINE | ID: mdl-30458075
10.
BMC Biotechnol ; 13: 64, 2013 Aug 07.
Article in English | MEDLINE | ID: mdl-23919313

ABSTRACT

BACKGROUND: Stable expression of transgenes is an important technique to analyze gene function. Various drug resistance genes, such as neo, pac, hph, zeo, bsd, and hisD, have been equally used as selection markers to isolate a transfectant without considering their dose-dependent characters. RESULTS: We quantitatively measured the variation of transgene expression levels in mouse embryonic stem (mES) cells, using a series of bi-cistronic expression vectors that contain Egfp expression cassette linked to each drug resistant gene via IRES with titration of the selective drugs, and found that the transgene expression levels achieved in each system with this vector design are in order, in which pac and zeo show sharp selection of transfectants with homogenously high expression levels. We also showed the importance of the choice of the drug selection system in gene-trap or gene targeting according to this order. CONCLUSIONS: The results of the present study clearly demonstrated that an appropriate choice of the drug resistance gene(s) is critical for a proper design of the experimental strategy.


Subject(s)
Embryonic Stem Cells/physiology , Gene Targeting/methods , Recombinant Fusion Proteins/analysis , Transgenes , Animals , Biotechnology/methods , Drug Resistance/genetics , Embryonic Stem Cells/metabolism , Genetic Markers/genetics , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Kinetics , Mice , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Selection, Genetic
11.
Proc Natl Acad Sci U S A ; 110(16): 6412-7, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23550161

ABSTRACT

Transcription factors (TFs) are able to regulate differentiation-related processes, including dedifferentiation and direct conversion, through the regulation of cell type-specific transcriptional profiles. However, the functional interactions between the TFs regulating different transcriptional profiles are not well understood. Here, we show that the TFs capable of inducing cell type-specific transcriptional profiles prevent the dedifferentiation induced by TFs for pluripotency. Of the large number of TFs expressed in a neural-lineage cell line, we identified a subset of TFs that, when overexpressed, strongly interfered with the dedifferentiation triggered by the procedure to generate induced pluripotent stem cells. This interference occurred through a maintenance mechanism of the cell type-specific transcriptional profile. Strikingly, the maintenance activity of the interfering TF set was strong enough to induce the cell line-specific transcriptional profile when overexpressed in a heterologous cell type. In addition, the TFs that interfered with dedifferentiation in hepatic-lineage cells involved TFs with known induction activity for hepatic-lineage cells. Our results suggest that dedifferentiation suppresses a cell type-specific transcriptional profile, which is primarily maintained by a small subset of TFs capable of inducing direct conversion. We anticipate that this functional correlation might be applicable in various cell types and might facilitate the identification of TFs with induction activity in efforts to understand differentiation.


Subject(s)
Cell Dedifferentiation/physiology , Gene Expression Regulation/physiology , Neurons/metabolism , Pluripotent Stem Cells/physiology , Transcription Factors/metabolism , Animals , Cell Line , Chromatin Immunoprecipitation , DNA Primers/genetics , Gene Expression Profiling , Hepatocytes/cytology , Mice , Microscopy, Electron, Transmission , Neurons/cytology , Oligonucleotide Array Sequence Analysis , Plasmids/genetics , RNA, Small Interfering/genetics
12.
J Biotechnol ; 154(4): 298-303, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21635926

ABSTRACT

Induction of a desired cell type by defined transcription factors (TFs) using iPS technology can be used for cell replacement therapy. However, to overcome problems such as tumor formation, genomic insertional mutagenesis by viral transduction in the induction process needs to be avoided using alternative approaches. One approach could be the direct delivery of TF protein by a protein transduction system, whereby a protein transduction domain (PTD) is fused to facilitate the penetration of cell membrane. However, fusion proteins, including TFs, are reported to be biologically less active through the interference of PTD with proper protein folding. Here, we report a proof-of-concept study in which TF proteins fused with PTDs could be reactivated by removal of PTDs from cells. We demonstrated that Sox2 and Oct3/4 proteins fused with PTD were less active in mouse embryonic stem cells. Removal of PTD by a site-specific protease, derived from tobacco etch virus (TEV), substantially restored the functionality of these proteins, proved by enhanced rescue ability for differentiation induced by endogenous Sox2 and Oct3/4 repression. These results suggest that, by removing a PTD inside the cells, directly delivered TF proteins may exert substantially enhanced function than presently considered.


Subject(s)
Transcription Factors/metabolism , Animals , Cell Line , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transcription Factors/genetics
13.
EMBO J ; 30(11): 2190-204, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21540835

ABSTRACT

Transcription factors and epigenetic modulators are involved in the maintenance of self-renewal in embryonic stem (ES) cells. Here, we demonstrate the existence of a regulatory loop in ES cells between Sox2, an indispensable transcription factor for self-renewal, and embryonic ectoderm development (Eed), an epigenetic modulator regulating histone methylation. We found that Sox2 and Eed positively regulate each other's expression. Interestingly, Sox2 overexpression suppressed the induction of differentiation-associated genes in Eed-deficient ES cells without restoring histone methylation. This Sox2-mediated suppression was prevented by knockdown of the histone acetyltransferase (HAT), Tip60 or Elp3, and Sox2 stimulated expression of these HATs. Furthermore, forced expression of either HAT resulted in repression of differentiation-associated genes in Eed-deficient cells. These results suggest that Sox2 overcame the phenotype of Eed-deficient ES cells by promoting histone acetylation. We also found that knockout of Eed and knockdown of these HATs synergistically enhanced the upregulation of differentiation-associated genes in ES cells. Taken together, our results suggest that the Eed/Sox2 regulatory loop contributes to the maintenance of self-renewal in ES cells by controlling histone methylation and acetylation.


Subject(s)
Embryonic Stem Cells/physiology , Gene Expression Regulation , Histones/metabolism , Repressor Proteins/biosynthesis , SOXB1 Transcription Factors/biosynthesis , Acetylation , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Methylation , Polycomb Repressive Complex 2 , Repressor Proteins/genetics , SOXB1 Transcription Factors/genetics
14.
J Biol Chem ; 286(13): 11593-603, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21296877

ABSTRACT

The prominent characteristics of pluripotent stem cells are their unique capacity to self-renew and pluripotency. Although pluripotent stem cell proliferation is maintained by specific intracellular phosphorylation signaling events, it has not been well characterized how the resulting phosphorylated proteins are subsequently regulated. We here report that the peptidylprolyl isomerase Pin1 is indispensable for the self-renewal and maintenance of pluripotent stem cells via the regulation of phosphorylated Oct4 and other substrates. Pin1 expression was found to be up-regulated upon the induction of induced pluripotent stem (iPS) cells, and the forced expression of Pin1 with defined reprogramming factors was observed to further enhance the frequency of iPS cell generation. The inhibition of Pin1 activity significantly suppressed colony formation and induced the aberrant differentiation of human iPS cells as well as murine ES cells. We further found that Pin1 interacts with the phosphorylated Ser(12)-Pro motif of Oct4 and that this in turn facilitates the stability and transcriptional activity functions of Oct4. Our current findings thus uncover an atypical role for Pin1 as a putative regulator of the induction and maintenance of pluripotency via the control of phosphorylation signaling. These data suggest that the manipulation of Pin1 function could be a potential strategy for the stable induction and proliferation of human iPS cells.


Subject(s)
Cell Proliferation , Induced Pluripotent Stem Cells/metabolism , Peptidylprolyl Isomerase/metabolism , Signal Transduction/physiology , Amino Acid Motifs , Cell Differentiation/physiology , Cell Line , Humans , Induced Pluripotent Stem Cells/cytology , NIMA-Interacting Peptidylprolyl Isomerase , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Peptidylprolyl Isomerase/genetics , Phosphorylation/physiology
15.
Int J Hematol ; 91(3): 360-72, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20157790

ABSTRACT

Embryonic stem (ES) cells are derived from blastocysts and are pluripotent. This pluripotency has attracted the interest of numerous researchers, both to expand our fundamental understanding of developmental biology and also because of potential applications in regenerative medicine. Systems biological studies have demonstrated that the pivotal transcription factors form a network. There they activate pluripotency-associated genes, including themselves, while repressing the developmentally regulated genes through co-occupation with various protein complexes. The chromatin structure characteristic of ES cells also contributes to the maintenance of the network. In this review, I focus on recent advances in our understanding of the transcriptional network that maintains pluripotency in mouse ES cells.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Animals , Mice , Transcription, Genetic/physiology
16.
Int J Dev Biol ; 54(4): 699-705, 2010.
Article in English | MEDLINE | ID: mdl-19757377

ABSTRACT

Insulin-dependent diabetes mellitus (IDDM) is characterized by the rapid development of potentially severe metabolic abnormalities resulting from insulin deficiency. The transplantation of insulin-producing cells is a promising approach for the treatment of IDDM. The transcription factor pancreatic duodenal homeobox 1 (Pdx1) plays an important role in the differentiation of pancreatic beta cells. In this study, the human Pdx1 gene was transduced and expressed in murine adipose tissue-derived stem cells (ASCs). To evaluate pancreatic repair, we used a mouse model of pancreatic damage resulting in hyperglycemia, which involves injection of mice with streptozotocin (STZ). STZ-treated mice transplanted with Pdx1-transduced ASCs (Pdx1-ASCs) showed significantly decreased blood glucose levels and increased survival, when compared with control mice. While stable expression of Pdx1 in ASCs did not induce the pancreatic phenotype in vitro in our experiment, the transplanted stem cells became engrafted in the pancreas, wherein they expressed insulin and C-peptide, which is a marker of insulin-producing cells. These results suggest that Pdx1-ASCs are stably engrafted in the pancreas, acquire a functional beta-cell phenotype, and partially restore pancreatic function in vivo. The ease and safety associated with extirpating high numbers of cells from adipose tissues support the applicability of this system to developing a new cell therapy for IDDM.


Subject(s)
Homeodomain Proteins/genetics , Hyperglycemia/metabolism , Insulin-Secreting Cells/metabolism , Insulin/biosynthesis , Stem Cells/cytology , Trans-Activators/genetics , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , C-Peptide/metabolism , Cell Differentiation/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/therapy , Humans , Insulin/genetics , Insulin/metabolism , Mice , Pancreas/cytology , Pancreas/metabolism , Stem Cell Transplantation , Stem Cells/metabolism , Streptozocin/metabolism
18.
Stem Cells ; 27(5): 1066-76, 2009 May.
Article in English | MEDLINE | ID: mdl-19418458

ABSTRACT

Stem cells have the remarkable ability to self-renew and to generate multiple cell types. Nucleostemin is one of proteins that are enriched in many types of stem cells. Targeted deletion of nucleostemin in the mouse results in developmental arrest at the implantation stage, indicating that nucleostemin is crucial for early embryogenesis. However, the molecular basis of nucleostemin function in early mouse embryos remains largely unknown, and the role of nucleostemin in tissue stem cells has not been examined by gene targeting analyses due to the early embryonic lethality of nucleostemin null animals. To address these questions, we generated inducible nucleostemin null embryonic stem (ES) cells in which both alleles of nucleostemin are disrupted, but nucleostemin cDNA under the control of a tetracycline-responsive transcriptional activator is introduced into the Rosa26 locus. We show that loss of nucleostemin results in reduced cell proliferation and increased apoptosis in both ES cells and ES cell-derived neural stem/progenitor cells. The reduction in cell viability is much more profound in ES cells than in neural stem/progenitor cells, an effect that is mediated at least in part by increased induction and accumulation of p53 and/or activated caspase-3 in ES cells than in neural stem/progenitor cells.


Subject(s)
Carrier Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/metabolism , Animals , Apoptosis/drug effects , Benzothiazoles/pharmacology , Biomarkers/metabolism , Carrier Proteins/genetics , Caspase 3/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Doxycycline/pharmacology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/enzymology , Enzyme Activation/drug effects , GTP-Binding Proteins , Gene Expression Regulation, Developmental/drug effects , Mice , Neurons/drug effects , Nuclear Proteins/genetics , Phenotype , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , RNA-Binding Proteins , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/metabolism
20.
In Vitro Cell Dev Biol Anim ; 45(1-2): 62-8, 2009.
Article in English | MEDLINE | ID: mdl-19037705

ABSTRACT

The transcription factor Pax2 is essential for kidney development in mice, and overexpression of Pax2 in chick embryos leads to ectopic formation of nephric structures. We have generated embryonic stem (ES) cell lines that repress Pax2 expression in a tetracycline-dependent manner. In the absence of tetracycline, embryoid bodies derived from these cell lines expressed Pax2 and subsequently integrin alpha8 and aquaporin-1 (Aqp1), both of which are possibly involved in kidney development. Considering the slow induction kinetics, our data suggest that Pax2 and additional factors that are induced in embryoid bodies synergistically regulate the two targets. The ES cell lines with inducible Pax2 expression will also be useful for dissecting genetic cascades functioning in a variety of organ development.


Subject(s)
Aquaporin 1/metabolism , Gene Expression , Integrin alpha Chains/metabolism , PAX2 Transcription Factor/metabolism , Up-Regulation , Animals , Cell Differentiation , Cell Line , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Mice , Protein Synthesis Inhibitors/pharmacology , Tetracycline/pharmacology , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...