Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Stem Cells ; 35(4): 872-885, 2017 04.
Article in English | MEDLINE | ID: mdl-27888558

ABSTRACT

Genomic aberrations have been identified in many human pluripotent stem cell (hPSC) cultures. Commonly observed duplications in portions of chromosomes 12p and 17q have been associated with increases in genetic instability and resistance to apoptosis, respectively. However, the phenotypic consequences related to sporadic mutations have not been evaluated to date. Here, we report on the effects of a single-copy deletion of the chr17p13.1 region, a sporadic mutation that spontaneously arose independently in several subclones of a human embryonic stem cell culture. Compared to cells with two normal copies of chr17p13.1 ("wild-type"), the cells with a single-copy deletion of this region ("mutant") displayed a selective advantage when exposed to stressful conditions, and retained a higher percentage of cells expressing the pluripotency marker POU5F1/OCT4 after 2 weeks of in vitro differentiation. Knockdown of TP53, which is a gene encompassed by the deleted region, in wild-type cells mimicked the chr17p13.1 deletion phenotype. Thus, sporadic mutations in hPSCs can have phenotypic effects that may impact their utility for clinical applications. Stem Cells 2017;35:872-885.


Subject(s)
Gene Dosage , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Mutation/genetics , Tumor Suppressor Protein p53/genetics , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromosomes, Human, Pair 17/genetics , Clone Cells , DNA Damage , DNA Repair/drug effects , Etoposide/pharmacology , Gene Expression Profiling , Gene Knockdown Techniques , Human Embryonic Stem Cells/drug effects , Humans , Phenotype , RNA, Small Interfering/metabolism , Staurosporine/pharmacology
2.
J Hepatol ; 64(6): 1315-26, 2016 06.
Article in English | MEDLINE | ID: mdl-26921690

ABSTRACT

BACKGROUND & AIMS: Hepatocytes differentiated from human embryonic stem cells (hESCs) have the potential to overcome the shortage of primary hepatocytes for clinical use and drug development. Many strategies for this process have been reported, but the functionality of the resulting cells is incomplete. We hypothesize that the functionality of hPSC-derived hepatocytes might be improved by making the differentiation method more similar to normal in vivo hepatic development. METHODS: We tested combinations of growth factors and small molecules targeting candidate signaling pathways culled from the literature to identify optimal conditions for differentiation of hESCs to hepatocytes, using qRT-PCR for stage-specific markers to identify the best conditions. Immunocytochemistry was then used to validate the selected conditions. Finally, induction of expression of metabolic enzymes in terminally differentiated cells was used to assess the functionality of the hESC-derived hepatocytes. RESULTS: Optimal differentiation of hESCs was attained using a 5-stage protocol. After initial induction of definitive endoderm (stage 1), we showed that inhibition of the WNT/ß-catenin pathway during the 2nd and 3rd stages of differentiation was required to specify first posterior foregut, and then hepatic gut cells. In contrast, during the 4th stage of differentiation, we found that activation of the WNT/ß-catenin pathway allowed generation of proliferative bipotent hepatoblasts, which then were efficiently differentiated into hepatocytes in the 5th stage by dual inhibition of TGF-ß and NOTCH signaling. CONCLUSION: Here, we show that stage-specific regulation of the WNT/ß-catenin pathway results in improved differentiation of hESCs to functional hepatocytes.


Subject(s)
Hepatocytes/cytology , Human Embryonic Stem Cells/cytology , Wnt Signaling Pathway/physiology , beta Catenin/physiology , Cell Differentiation , Cells, Cultured , Cytochrome P-450 Enzyme System/metabolism , Humans , Receptors, Notch/physiology , Serum Albumin, Human/analysis , Transforming Growth Factor beta/antagonists & inhibitors , alpha-Fetoproteins/analysis
3.
Nat Protoc ; 8(2): 430-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23424751

ABSTRACT

Large-scale production of hepatocytes from a variety of genetic backgrounds would be beneficial for drug screening and to provide a source of cells to be used as a substitute for liver transplantation. However, fully functional primary hepatocytes remain difficult to expand in vitro, and circumventing this problem by using an alternative source of cells is desirable. Here we describe a 25-d protocol to direct the differentiation of human pluripotent stem cells into a near-homogenous population of hepatocyte-like cells. As cells progress through this protocol, they express genes in a chronological manner similar to that described during in vivo hepatic development. The protocol relies on culture systems devoid of serum, feeders or complex extracellular matrices, which enable molecular analyses without interference from unknown factors. This approach works efficiently with human embryonic stem cells and human induced pluripotent stem cells and was recently used to model liver diseases in vitro.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/physiology , Hepatocytes/cytology , Pluripotent Stem Cells/cytology , Humans , Serum Albumin/metabolism , Serum Albumin, Human , Transcriptome , alpha 1-Antitrypsin/metabolism
4.
Stem Cells Dev ; 21(2): 191-205, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-21699412

ABSTRACT

Lineage reporters of human embryonic stem cell (hESC) lines are useful for differentiation studies and drug screening. Previously, we created reporter lines driven by an elongation factor 1 alpha (EF1α) promoter at a chromosome 13q32.3 locus in the hESC line WA09 and an abnormal hESC line BG01V in a site-specific manner. Expression of reporters in these lines was maintained in long-term culture at undifferentiated state. However, when these cells were differentiated into specific lineages, reduction in reporter expression was observed, indicating transgene silencing. To develop an efficient and reliable genetic engineering strategy in hESCs, we used chromatin insulator elements to flank single-copy transgenes and integrated the combined expression constructs via PhiC31/R4 integrase-mediated recombination technology to the chromosome 13 locus precisely. Two copies of cHS4 double-insulator sequences were placed adjacent to both 5' and 3' of the promoter reporter constructs. The green fluorescent protein (GFP) gene was driven by EF1α or CMV early enhancer/chicken ß actin (CAG) promoter. In the engineered hESC lines, for both insulated CAG-GFP and EF1α-GFP, constitutive expression at the chromosome 13 locus was maintained during prolonged culture and in directed differentiation assays toward diverse types of neurons, pancreatic endoderm, and mesodermal progeny. In particular, described here is the first normal hESC fluorescent reporter line that robustly expresses GFP in both the undifferentiated state and throughout dopaminergic lineage differentiation. The dual strategy of utilizing insulator sequences and integration at the constitutive chromosome 13 locus ensures appropriate transgene expression. This is a valuable tool for lineage development study, gain- and loss-of-function experiments, and human disease modeling using hESCs.


Subject(s)
Chromatin/genetics , Embryonic Stem Cells , Genetic Engineering/methods , Insulator Elements/genetics , Recombination, Genetic , Cell Differentiation , Cell Line, Transformed , Cell Lineage , Chromosomes, Human, Pair 13 , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genes, Reporter , Genetic Loci , Genetic Vectors , Green Fluorescent Proteins/genetics , Humans , Integrases/genetics , Integrases/metabolism , Peptide Elongation Factor 1/genetics , Promoter Regions, Genetic , Transgenes
5.
Med Sci (Paris) ; 26(12): 1061-6, 2010 Dec.
Article in French | MEDLINE | ID: mdl-21187045

ABSTRACT

Hepatocyte transplantation is considered as an alternative to organ transplantation in particular for the treatment of liver metabolic diseases. However, due to the difficulties to obtain a large number of hepatocytes, new sources of cells are needed. These cells could be either of hepatic origin (hepatic stem cells) or extrahepatic such as mesenchymal stem cells or pluripotent stem cells (human embryonic stem cells [hESC] or iPS). We developed a new method to differentiate hESCs into fetal hepatocytes. These conditions recapitulate the main liver developmental stages, using fully defined medium devoid of animal products or unknown factors. The differentiated cells express many fetal hepatocytes markers (cytochrome P450 3A7, albumin, alpha-1-antitrypsin, etc.). The cells display specific hepatic functions (ammonia metabolism, excretion of indocyanin green) and are capable to engraft and express hepatic proteins two months after transplantation into newborn uPAxrag2gc-/- mouse liver. We have also showed that this approach is transposable to human iPS, and further studies on animal models will allow us to compare the in vivo potential of these two sources of pluripotent cells. Finally, only studies on large animals such as nonhuman primates will validate an eventual clinical application.


Subject(s)
Embryonic Stem Cells/cytology , Hepatocytes/cytology , Pluripotent Stem Cells/cytology , Animals , Animals, Newborn , Biomarkers , Cell Differentiation , Cell Lineage , Cell Transdifferentiation , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/transplantation , Culture Media/pharmacology , Digestive System/embryology , Embryonic Stem Cells/drug effects , Endoderm/cytology , Gene Expression Regulation, Developmental , Hepatocytes/metabolism , Hepatocytes/transplantation , Humans , Liver/cytology , Liver/embryology , Mice , Pluripotent Stem Cells/transplantation , Transplantation, Heterologous
6.
Methods Mol Biol ; 640: 41-55, 2010.
Article in English | MEDLINE | ID: mdl-20645045

ABSTRACT

Hepatocyte transplantation has become an alternative to orthotopic liver transplantation for the treatment of liver metabolic diseases. However, there is an increasing lack of donor organs and isolated mature hepatocytes are difficult to manipulate and cannot be expanded in vitro. It is therefore necessary to find alternative sources of hepatocytes, and different approaches to evaluate the therapeutic potential of stem cells of different origins are being developed. Hepatic progenitors (hepatoblasts) and/or foetal hepatocytes isolated from foetal livers may be one potential source to generate fully differentiated hepatocytes. We have reported that human foetal liver cells can be isolated and cultured. These cells also engraft and differentiate into mature hepatocytes in situ after transplantation into immunodeficient mice. Foetal cell populations could also be used as targets for gene therapy since efficient gene transfer is achieved with retroviral vectors. Use of such experimental approaches will help design strategies for clinical applications of liver cell therapy with hepatic progenitors.


Subject(s)
Cell Separation/methods , Fetus/cytology , Hepatocytes/cytology , Hepatocytes/transplantation , Animals , Cell Differentiation , Cells, Cultured , Female , Genetic Vectors/genetics , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Lentivirus/genetics , Mice , Mice, SCID , Pregnancy , Transduction, Genetic
7.
Hepatology ; 51(5): 1754-65, 2010 May.
Article in English | MEDLINE | ID: mdl-20301097

ABSTRACT

UNLABELLED: Generation of hepatocytes from human embryonic stem cells (hESCs) could represent an advantageous source of cells for cell therapy approaches as an alternative to orthotopic liver transplantation. However, the generation of differentiated hepatocytes from hESCs remains a major challenge, especially using a method compatible with clinical applications. We report a novel approach to differentiate hESCs into functional hepatic cells using fully defined culture conditions, which recapitulate essential stages of liver development. hESCs were first differentiated into a homogenous population of endoderm cells using a combination of activin, fibroblast growth factor 2, and bone morphogenetic protein 4 together with phosphoinositide 3-kinase inhibition. The endoderm cells were then induced to differentiate further into hepatic progenitors using fibroblast growth factor 10, retinoic acid, and an inhibitor of activin/nodal receptor. After further maturation, these cells expressed markers of mature hepatocytes, including asialoglycoprotein receptor, tyrosine aminotransferase, alpha1-antitrypsin, Cyp7A1, and hepatic transcription factors such as hepatocyte nuclear factors 4alpha and 6. Furthermore, the cells generated under these conditions exhibited hepatic functions in vitro, including glycogen storage, cytochrome activity, and low-density lipoprotein uptake. After transduction with a green fluorescent protein-expressing lentivector and transplantation into immunodeficient uPA transgenic mice, differentiated cells engrafted into the liver, grew, and expressed human albumin and alpha1-antitrypsin as well as green fluorescent protein for at least 8 weeks. In addition, we showed that hepatic cells could be generated from human-induced pluripotent cells derived from reprogrammed fibroblasts, demonstrating the efficacy of this approach with pluripotent stem cells of diverse origins. CONCLUSION: We have developed a robust and efficient method to differentiate pluripotent stem cells into hepatic cells, which exhibit characteristics of human hepatocytes. Our approach should facilitate the development of clinical grade hepatocytes for transplantation and for research on drug discovery.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Hepatocytes/cytology , Liver/embryology , Activins/pharmacology , Animals , Benzamides/pharmacology , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/physiology , Chromones/pharmacology , Dioxoles/pharmacology , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Developmental/drug effects , Humans , Mice , Morpholines/pharmacology , Pluripotent Stem Cells/cytology , Tretinoin/pharmacology
8.
Stem Cells ; 27(11): 2655-66, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19688839

ABSTRACT

Human pluripotent stem cells from embryonic origins and those generated from reprogrammed somatic cells share many characteristics, including indefinite proliferation and a sustained capacity to differentiate into a wide variety of cell types. However, it remains to be demonstrated whether both cell types rely on similar mechanisms to maintain their pluripotent status and to control their differentiation. Any differences in such mechanisms would suggest that reprogramming of fibroblasts to generate induced pluripotent stem cells (iPSCs) results in novel states of pluripotency. In that event, current methods for expanding and differentiating human embryonic stem cells (ESCs) might not be directly applicable to human iPSCs. However, we show here that human iPSCs rely on activin/nodal signaling to control Nanog expression and thereby maintain pluripotency, thus revealing their mechanistic similarity to human ESCs. We also show that growth factors necessary and sufficient for achieving specification of human ESCs into extraembryonic tissues, neuroectoderm, and mesendoderm also drive differentiation of human iPSCs into the same tissues. Importantly, these experiments were performed in fully chemically defined medium devoid of factors that could obscure analysis of developmental mechanisms or render the resulting tissues incompatible with future clinical applications. Together these data reveal that human iPSCs rely on mechanisms similar to human ESCs to maintain their pluripotency and to control their differentiation, showing that these pluripotent cell types are functionally equivalent.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Signal Transduction/physiology , Activin Receptors/antagonists & inhibitors , Activins/pharmacology , Adult , Animals , Benzamides/pharmacology , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Culture Media , Dioxoles/pharmacology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Female , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/cytology , Flow Cytometry , Fluorescent Antibody Technique , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/physiology , Male , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/physiology , Polymerase Chain Reaction , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/physiology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/physiology , Signal Transduction/drug effects , Signal Transduction/genetics
9.
PLoS One ; 4(6): e6082, 2009 Jun 30.
Article in English | MEDLINE | ID: mdl-19564924

ABSTRACT

Human embryonic stem cells have unique value for regenerative medicine, as they are capable of differentiating into a broad variety of cell types. Therefore, defining the signalling pathways that control early cell fate decisions of pluripotent stem cells represents a major task. Moreover, modelling the early steps of embryonic development in vitro may provide the best approach to produce cell types with native properties. Here, we analysed the function of key developmental growth factors such as Activin, FGF and BMP in the control of early cell fate decisions of human pluripotent stem cells. This analysis resulted in the development and validation of chemically defined culture conditions for achieving specification of human embryonic stem cells into neuroectoderm, mesendoderm and into extra-embryonic tissues. Importantly, these defined culture conditions are devoid of factors that could obscure analysis of developmental mechanisms or render the resulting tissues incompatible with future clinical applications. Importantly, the growth factor roles defined using these culture conditions similarly drove differentiation of mouse epiblast stem cells derived from post implantation embryos, thereby reinforcing the hypothesis that epiblast stem cells share a common embryonic identity with human pluripotent stem cells. Therefore the defined growth factor conditions described here represent an essential step toward the production of mature cell types from pluripotent stem cells in conditions fully compatible with clinical use ant also provide a general approach for modelling the early steps of mammalian embryonic development.


Subject(s)
Cell Differentiation , Cell Lineage , Embryonic Stem Cells/cytology , Germ Layers/cytology , Stem Cells/cytology , Animals , Bone Morphogenetic Protein 4/metabolism , Ectoderm/metabolism , Embryonic Stem Cells/physiology , Endoderm/metabolism , Humans , Mesoderm/metabolism , Mice , Models, Biological , Signal Transduction
10.
Exp Cell Res ; 315(19): 3396-405, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19615360

ABSTRACT

UNLABELLED: The success of hepatocyte transplantation has been limited by the low efficiency of transplanted cell integration into liver parenchyma. Human fetal hepatic progenitor cells (hepatoblasts) engraft more effectively than adult hepatocytes in mouse livers. However, the signals required for their integration are not yet fully understood. We investigated the role of HGF on the migration and invasive ability of human hepatic progenitors in vitro and in vivo. Hepatoblasts were isolated from the livers of human fetuses between 10 and 12 weeks of gestation. Their invasive ability was assessed in the presence or absence of HGF. These cells were also transplanted into immunodeficient mice and analyzed by immunohistochemistry. In contrast to TNF-alpha, HGF increased the motogenesis and invasiveness of hepatoblasts, but not of human adult hepatocytes, via phosphorylation of extracellular signal-regulated kinase (ERK) 1/2. The invasive ability of human hepatoblasts correlated with the expression and secretion of matrix metalloproteinases (MMPs). Hepatoblasts stimulated with HGF prior transplantation into newborn mice migrated from the portal area into the hepatic parenchyma. CONCLUSIONS: In contrast to adult hepatocytes, hepatoblasts display invasive ability that can be modulated by HGF in vitro and in vivo.


Subject(s)
Cell Movement , Hepatocyte Growth Factor/physiology , Hepatocytes/transplantation , Stem Cell Transplantation/methods , Stem Cells/physiology , Animals , Cell Proliferation , Fetus , Hepatocyte Growth Factor/pharmacology , Humans , Mice , Mice, SCID , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Stem Cells/cytology , Transplantation, Heterologous
11.
Mol Cell Biol ; 25(2): 854-64, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15632084

ABSTRACT

Mitogen-activated protein (MAP) kinase phosphatases (MKPs) are dual-specificity phosphatases that dephosphorylate phosphothreonine and phosphotyrosine residues within MAP kinases. Here, we describe a novel posttranslational mechanism for regulating MKP-3/Pyst1/DUSP6, a member of the MKP family that is highly specific for extracellular signal-regulated kinase 1 and 2 (ERK1/2) inactivation. Using a fibroblast model in which the expression of either MKP-3 or a more stable MKP-3-green fluorescent protein (GFP) chimera was induced by tetracycline, we found that serum induces the phosphorylation of MKP-3 and its subsequent degradation by the proteasome in a MEK1 and MEK2 (MEK1/2)-ERK1/2-dependent manner. In vitro phosphorylation assays using glutathione S-transferase (GST)-MKP-3 fusion proteins indicated that ERK2 could phosphorylate MKP-3 on serines 159 and 197. Tetracycline-inducible cell clones expressing either single or double serine mutants of MKP-3 or MKP-3-GFP confirmed that these two sites are targeted by the MEK1/2-ERK1/2 module in vivo. Double serine mutants of MKP-3 or MKP-3-GFP were more efficiently protected from degradation than single mutants or wild-type MKP-3, indicating that phosphorylation of either serine by ERK1/2 enhances proteasomal degradation of MKP-3. Hence, double mutation caused a threefold increase in the half-life of MKP-3. Finally, we show that the phosphorylation of MKP-3 has no effect on its catalytic activity. Thus, ERK1/2 exert a positive feedback loop on their own activity by promoting the degradation of MKP-3, one of their major inactivators in the cytosol, a situation opposite to that described for the nuclear phosphatase MKP-1.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Tyrosine Phosphatases/metabolism , Serine/metabolism , Animals , Cell Line , Dual Specificity Phosphatase 6 , Enzyme Stability , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Mutagenesis, Site-Directed , Phosphorylation , Protein Tyrosine Phosphatases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...