Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Aliment Pharmacol Ther ; 51(12): 1305-1320, 2020 06.
Article in English | MEDLINE | ID: mdl-32383295

ABSTRACT

BACKGROUND: Non-alcoholic steatohepatitis (NASH) is a severe form of non-alcoholic fatty liver disease (NAFLD) characterised by liver fat accumulation, inflammation and progressive fibrosis. Emerging data indicate that genetic susceptibility increases risks of NAFLD, NASH and NASH-related cirrhosis. AIMS: To review NASH genetics and discuss the potential for precision medicine approaches to treatment. METHOD: PubMed search and inclusion of relevant literature. RESULTS: Single-nucleotide polymorphisms in PNPLA3, TM6SF2, GCKR, MBOAT7 and HSD17B13 are clearly associated with NASH development or progression. These genetic variants are common and have moderate-to-large effect sizes for development of NAFLD, NASH and hepatocellular carcinoma (HCC). The genes play roles in lipid remodelling in lipid droplets, hepatic very low-density lipoprotein (VLDL) secretion and de novo lipogenesis. The PNPLA3 I148M variant (rs738409) has large effects, with approximately twofold increased odds of NAFLD and threefold increased odds of NASH and HCC per allele. Obesity interacts with PNPLA3 I148M to elevate liver fat content and increase rates of NASH. Although the isoleucine-to-methionine substitution at amino acid position 148 of the PNPLA3 enzyme inactivates its lipid remodelling activity, the effect of PNPLA3 I148M results from trans-repression of another lipase (ATGL/PNPLA2) by sequestration of a shared cofactor (CGI-58/ABHD5), leading to decreased hepatic lipolysis and VLDL secretion. In homozygous Pnpla3 I148M knock-in rodent models of NAFLD, targeted PNPLA3 mRNA knockdown reduces hepatic steatosis, inflammation and fibrosis. CONCLUSION: The emerging genetic and molecular understanding of NASH paves the way for novel interventions, including precision medicines that can modulate the activity of specific genes associated with NASH.


Subject(s)
Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/therapy , Precision Medicine/trends , Alleles , Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/therapy , Disease Progression , Genetic Predisposition to Disease , Genetic Testing/methods , Genetic Testing/trends , Humans , Liver Neoplasms/complications , Liver Neoplasms/genetics , Liver Neoplasms/therapy , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/pathology , Polymorphism, Single Nucleotide , Precision Medicine/methods , Prognosis
2.
Stem Cell Rev Rep ; 15(1): 13-22, 2019 02.
Article in English | MEDLINE | ID: mdl-30343468

ABSTRACT

The global burden of diabetes has drastically increased over the past decades and in 2017 approximately 4 million deaths were caused by diabetes and cardiovascular complications. Diabetic cardiomyopathy is a common complication of diabetes with early manifestations of diastolic dysfunction and left ventricular hypertrophy with subsequent progression to systolic dysfunction and ultimately heart failure. An in vitro model accurately recapitulating key processes of diabetic cardiomyopathy would provide a useful tool for investigations of underlying disease mechanisms to further our understanding of the disease and thereby potentially advance treatment strategies for patients. With their proliferative capacity and differentiation potential, human induced pluripotent stem cells (iPSCs) represent an appealing cell source for such a model system and cardiomyocytes derived from induced pluripotent stem cells have been used to establish other cardiovascular related disease models. Here we review recently made advances and discuss challenges still to be overcome with regard to diabetic cardiomyopathy models, with a special focus on iPSC-based systems. Recent publications as well as preliminary data presented here demonstrate the feasibility of generating cardiomyocytes with a diabetic phenotype, displaying insulin resistance, impaired calcium handling and hypertrophy. However, capturing the full metabolic- and functional phenotype of the diabetic cardiomyocyte remains to be accomplished.


Subject(s)
Diabetic Cardiomyopathies/pathology , Induced Pluripotent Stem Cells/cytology , Models, Biological , Myocytes, Cardiac/cytology , Animals , Diabetic Cardiomyopathies/genetics , Disease Models, Animal , Humans
3.
Stem Cells ; 36(12): 1816-1827, 2018 12.
Article in English | MEDLINE | ID: mdl-30171748

ABSTRACT

Cell-based models of the blood-brain barrier (BBB) are important for increasing the knowledge of BBB formation, degradation and brain exposure of drug substances. Human models are preferred over animal models because of interspecies differences in BBB structure and function. However, access to human primary BBB tissue is limited and has shown degeneration of BBB functions in vitro. Human induced pluripotent stem cells (iPSCs) can be used to generate relevant cell types to model the BBB with human tissue. We generated a human iPSC-derived model of the BBB that includes endothelial cells in coculture with pericytes, astrocytes and neurons. Evaluation of barrier properties showed that the endothelial cells in our coculture model have high transendothelial electrical resistance, functional efflux and ability to discriminate between CNS permeable and non-permeable substances. Whole genome expression profiling revealed transcriptional changes that occur in coculture, including upregulation of tight junction proteins, such as claudins and neurotransmitter transporters. Pathway analysis implicated changes in the WNT, TNF, and PI3K-Akt pathways upon coculture. Our data suggest that coculture of iPSC-derived endothelial cells promotes barrier formation on a functional and transcriptional level. The information about gene expression changes in coculture can be used to further improve iPSC-derived BBB models through selective pathway manipulation. Stem Cells 2018;36:1816-12.


Subject(s)
Blood-Brain Barrier/metabolism , Induced Pluripotent Stem Cells/metabolism , Transcriptome/physiology , Cell Differentiation , Humans
4.
Stem Cell Reports ; 10(3): 1030-1045, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29456185

ABSTRACT

In vivo studies of human brain cellular function face challenging ethical and practical difficulties. Animal models are typically used but display distinct cellular differences. One specific example is astrocytes, recently recognized for contribution to neurological diseases and a link to the genetic risk factor apolipoprotein E (APOE). Current astrocytic in vitro models are questioned for lack of biological characterization. Here, we report human induced pluripotent stem cell (hiPSC)-derived astroglia (NES-Astro) developed under defined conditions through long-term neuroepithelial-like stem (ltNES) cells. We characterized NES-Astro and astrocytic models from primary sources, astrocytoma (CCF-STTG1), and hiPSCs through transcriptomics, proteomics, glutamate uptake, inflammatory competence, calcium signaling response, and APOE secretion. Finally, we assess modulation of astrocyte biology using APOE-annotated compounds, confirming hits of the cholesterol biosynthesis pathway in adult and hiPSC-derived astrocytes. Our data show large diversity among astrocytic models and emphasize a cellular context when studying astrocyte biology.


Subject(s)
Astrocytes/physiology , Induced Pluripotent Stem Cells/physiology , Neural Stem Cells/physiology , Neurons/physiology , Apolipoproteins E/metabolism , Astrocytes/metabolism , Brain/metabolism , Brain/physiology , Cell Differentiation/physiology , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism
5.
Stem Cell Res ; 20: 105-114, 2017 04.
Article in English | MEDLINE | ID: mdl-28334554

ABSTRACT

A fast track "Hot Start" process was implemented to launch the European Bank for Induced Pluripotent Stem Cells (EBiSC) to provide early release of a range of established control and disease linked human induced pluripotent stem cell (hiPSC) lines. Established practice amongst consortium members was surveyed to arrive at harmonised and publically accessible Standard Operations Procedures (SOPs) for tissue procurement, bio-sample tracking, iPSC expansion, cryopreservation, qualification and distribution to the research community. These were implemented to create a quality managed foundational collection of lines and associated data made available for distribution. Here we report on the successful outcome of this experience and work flow for banking and facilitating access to an otherwise disparate European resource, with lessons to benefit the international research community. ETOC: The report focuses on the EBiSC experience of rapidly establishing an operational capacity to procure, bank and distribute a foundational collection of established hiPSC lines. It validates the feasibility and defines the challenges of harnessing and integrating the capability and productivity of centres across Europe using commonly available resources currently in the field.


Subject(s)
Biological Specimen Banks , Induced Pluripotent Stem Cells/cytology , Cell Line , Cryopreservation , Europe , Humans
6.
Physiol Genomics ; 48(11): 771-784, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27591124

ABSTRACT

Regenerative therapies hold great potential to change the treatment paradigm for cardiac diseases. Human cardiac progenitor cells can be used for drug discovery in this area and also provide a renewable source of cardiomyocytes. However, a better understanding of their characteristics is critical for interpreting data obtained from drug screening using these cells. In the present study, we performed global transcriptional analysis of two important sources of cardiac progenitors, i.e., patient epicardium-derived cells (EPDCs) and cardiac progenitor cells (CPCs) derived from human induced pluripotent stem cells. In addition, we also compared the gene expression profiles of these cells when they were cultured under normoxic and hypoxic conditions. We identified 3,289 mRNAs that were differentially expressed between EPDCs and CPCs. Gene ontology annotation and pathway enrichment analyses further revealed possible unique functions of these two cell populations. Notably, the impact of hypoxia vs normoxia on gene expression was modest and only a few genes (e.g., AK4, ALDOC, BNIP3P1, PGK1, and SLC2A1) were upregulated in EPDCs and CPCs after the cells were exposed to low oxygen for 24 h. Finally, we also performed a focused analysis of the gene expression patterns of a predefined set of 92 paracrine factors. We identified 30 of these genes as differentially expressed, and 29 were expressed at higher levels in EPDCs compared with CPCs. Taken together, the results of the present study advance our understanding of the transcriptional programs in EPDCs and CPCs and highlights important differences and similarities between these cell populations.


Subject(s)
Gene Expression Profiling , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Pericardium/cytology , Biomarkers/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cluster Analysis , Gene Expression Regulation/drug effects , Gene Ontology , Humans , Induced Pluripotent Stem Cells/drug effects , Molecular Sequence Annotation , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxygen/pharmacology , Paracrine Communication/genetics , Protein Interaction Maps/drug effects , Protein Interaction Maps/genetics
7.
Stem Cells Transl Med ; 5(2): 164-74, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26683871

ABSTRACT

Several progenitor cell populations have been reported to exist in hearts that play a role in cardiac turnover and/or repair. Despite the presence of cardiac stem and progenitor cells within the myocardium, functional repair of the heart after injury is inadequate. Identification of the signaling pathways involved in the expansion and differentiation of cardiac progenitor cells (CPCs) will broaden insight into the fundamental mechanisms playing a role in cardiac homeostasis and disease and might provide strategies for in vivo regenerative therapies. To understand and exploit cardiac ontogeny for drug discovery efforts, we developed an in vitro human induced pluripotent stem cell-derived CPC model system using a highly enriched population of KDR(pos)/CKIT(neg)/NKX2.5(pos) CPCs. Using this model system, these CPCs were capable of generating highly enriched cultures of cardiomyocytes under directed differentiation conditions. In order to facilitate the identification of pathways and targets involved in proliferation and differentiation of resident CPCs, we developed phenotypic screening assays. Screening paradigms for therapeutic applications require a robust, scalable, and consistent methodology. In the present study, we have demonstrated the suitability of these cells for medium to high-throughput screens to assess both proliferation and multilineage differentiation. Using this CPC model system and a small directed compound set, we identified activin-like kinase 5 (transforming growth factor-ß type 1 receptor kinase) inhibitors as novel and potent inducers of human CPC differentiation to cardiomyocytes. Significance: Cardiac disease is a leading cause of morbidity and mortality, with no treatment available that can result in functional repair. This study demonstrates how differentiation of induced pluripotent stem cells can be used to identify and isolate cell populations of interest that can translate to the adult human heart. Two separate examples of phenotypic screens are discussed, demonstrating the value of this biologically relevant and reproducible technology. In addition, this assay system was able to identify novel and potent inducers of differentiation and proliferation of induced pluripotent stem cell-derived cardiac progenitor cells.


Subject(s)
Cell Differentiation/drug effects , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/pharmacology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Biomarkers/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Expression , High-Throughput Screening Assays , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phenotype , Plasmids/chemistry , Plasmids/metabolism , Primary Cell Culture , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Small Molecule Libraries/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
8.
PLoS One ; 9(11): e113620, 2014.
Article in English | MEDLINE | ID: mdl-25419971

ABSTRACT

With accelerating rates of obesity and type 2 diabetes world-wide, interest in studying the adipocyte and adipose tissue is increasing. Human adipose derived stem cells--differentiated to adipocytes in vitro--are frequently used as a model system for white adipocytes, as most of their pathways and functions resemble mature adipocytes in vivo. However, these cells are not completely like in vivo mature adipocytes. Hosting the cells in a more physiologically relevant environment compared to conventional two-dimensional cell culturing on plastic surfaces, can produce spatial cues that drive the cells towards a more mature state. We investigated the adipogenesis of adipose derived stem cells on electro spun polycaprolactone matrices and compared functionality to conventional two-dimensional cultures as well as to human primary mature adipocytes. To assess the degree of adipogenesis we measured cellular glucose-uptake and lipolysis and used a range of different methods to evaluate lipid accumulation. We compared the averaged results from a whole population with the single cell characteristics--studied by coherent anti-Stokes Raman scattering microscopy--to gain a comprehensive picture of the cell phenotypes. In adipose derived stem cells differentiated on a polycaprolactone-fiber matrix; an increased sensitivity in insulin-stimulated glucose uptake was detected when cells were grown on either aligned or random matrices. Furthermore, comparing differentiation of adipose derived stem cells on aligned polycaprolactone-fiber matrixes, to those differentiated in two-dimensional cultures showed, an increase in the cellular lipid accumulation, and hormone sensitive lipase content. In conclusion, we propose an adipocyte cell model created by differentiation of adipose derived stem cells on aligned polycaprolactone-fiber matrices which demonstrates increased maturity, compared to 2D cultured cells.


Subject(s)
Adipogenesis , Adipose Tissue/cytology , Polyesters , Stem Cells/cytology , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue/metabolism , Adult , Cell Culture Techniques/methods , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Female , Gene Expression , Glucose/metabolism , Glucose/pharmacokinetics , Humans , Lipids/analysis , Lipolysis , Male , Microscopy, Fluorescence, Multiphoton , Reverse Transcriptase Polymerase Chain Reaction , Spectrum Analysis, Raman/methods , Stem Cells/metabolism , Tissue Scaffolds/chemistry , Young Adult
9.
Arch Toxicol ; 88(7): 1427-37, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24912781

ABSTRACT

Human-induced pluripotent stem cell-derived hepatocytes (hiPSC-Hep) hold great potential as an unlimited cell source for toxicity testing in drug discovery research. However, little is known about mechanisms of compound toxicity in hiPSC-Hep. In this study, modified mRNA was used to reprogram foreskin fibroblasts into hiPSC that were differentiated into hiPSC-Hep. The hiPSC-Hep expressed characteristic hepatic proteins and exhibited cytochrome P450 (CYP) enzyme activities. Next, the hiPSC-Hep, primary cryopreserved human hepatocytes (cryo-hHep) and the hepatic cell lines HepaRG and Huh7 were treated with staurosporine and acetaminophen, and the toxic responses were compared. In addition, the expression of genes regulating and executing apoptosis was analyzed in the different cell types. Staurosporine, an inducer of apoptosis, decreased ATP levels and activated caspases 3 and 7 in all cell types, but to less extent in Huh7. Furthermore, a hierarchical clustering and a principal component analysis (PCA) of the expression of apoptosis-associated genes separated cryo-hHep from the other cell types, while an enrichment analysis of apoptotic pathways identified hiPSC-Hep as more similar to cryo-hHep than the hepatic cell lines. Finally, acetaminophen induced apoptosis in hiPSC-Hep, HepaRG and Huh7, while the compound initiated a direct necrotic response in cryo-hHep. Our results indicate that for studying compounds initiating apoptosis directly hiPSC-Hep may be a good alternative to cryo-hHep. Furthermore, for compounds with more complex mechanisms of toxicity involving metabolic activation, such as acetaminophen, our data suggest that the cause of cell death depends on a balance between factors controlling death signals and the drug-metabolizing capacity.


Subject(s)
Acetaminophen/toxicity , Hepatocytes/drug effects , Staurosporine/toxicity , Toxicity Tests/methods , Acetaminophen/metabolism , Apoptosis/drug effects , Cell Differentiation , Cell Line , Cells, Cultured , Chemical and Drug Induced Liver Injury/diagnosis , Chemical and Drug Induced Liver Injury/etiology , Cryopreservation/methods , Fibroblasts/cytology , Foreskin , Hepatocytes/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Male , Principal Component Analysis
10.
Biochem Pharmacol ; 86(5): 691-702, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23856292

ABSTRACT

Human embryonic and induced pluripotent stem cell-derived hepatocytes (hESC-Hep and hiPSC-Hep) have the potential to provide relevant human in vitro model systems for toxicity testing and drug discovery studies. In this study, the expression and function of important drug metabolizing cytochrome P450 (CYP) enzymes and transporter proteins in hESC-Hep and hiPSC-Hep were compared to cryopreserved human primary hepatocytes (hphep) and HepG2 cells. Overall, CYP activities in hESC-Hep and hiPSC-Hep were much lower than in hphep cultured for 4 h, but CYP1A and 3A activities were comparable to levels in hphep cultured for 48h (CYP1A: 35% and 26% of 48 h hphep, respectively; CYP3A: 80% and 440% of 48 h hphep, respectively). Importantly, in hESC-Hep and hiPSC-Hep, CYP activities were stable or increasing for at least one week in culture which was in contrast to the rapid loss of CYP activities in cultured hphep between 4 and 48 h after plating. With regard to transporters, in hESC-Hep and hiPSC-Hep, pronounced NTCP activity (17% and 29% of 4 h hphep, respectively) and moderate BSEP activity (6% and 8% of 4 h hphep, respectively) were observed. Analyses of mRNA expression and immunocytochemistry supported the observed CYP and transporter activities and showed expression of additional CYPs and transporters. In conclusion, the stable expression and function of CYPs and transporters in hESC-Hep and hiPSC-Hep for at least one week opens up the possibility to reproducibly perform long term and extensive studies, e.g. chronic toxicity testing, in a stem cell-derived hepatic system.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Embryonic Stem Cells/metabolism , Hepatocytes/metabolism , Induced Pluripotent Stem Cells/metabolism , Membrane Transport Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Cell Line , Cytochrome P-450 Enzyme System/genetics , Hepatocytes/enzymology , Humans , Membrane Transport Proteins/genetics , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Organic Cation Transporter 1/genetics , Organic Cation Transporter 1/metabolism , Real-Time Polymerase Chain Reaction
11.
Carcinogenesis ; 34(6): 1393-402, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23393228

ABSTRACT

As the conventional approach to assess the potential of a chemical to cause cancer in humans still includes the 2-year rodent carcinogenicity bioassay, development of alternative methodologies is needed. In the present study, the transcriptomics responses following exposure to genotoxic (GTX) and non-genotoxic (NGTX) hepatocarcinogens and non-carcinogens (NC) in five liver-based in vitro models, namely conventional and epigenetically stabilized cultures of primary rat hepatocytes, the human hepatoma-derived cell lines HepaRG and HepG2 and human embryonic stem cell-derived hepatocyte-like cells, are examined. For full characterization of the systems, several bioinformatics approaches are employed including gene-based, ConsensusPathDB-based and classification analysis. They provide convincingly similar outcomes, namely that upon exposure to carcinogens, the HepaRG generates a gene classifier (a gene classifier is defined as a selected set of characteristic gene signatures capable of distinguishing GTX, NGTX carcinogens and NC) able to discriminate the GTX carcinogens from the NGTX carcinogens and NC. The other in vitro models also yield cancer-relevant characteristic gene groups for the GTX exposure, but some genes are also deregulated by the NGTX carcinogens and NC. Irrespective of the tested in vitro model, the most uniformly expressed pathways following GTX exposure are the p53 and those that are subsequently induced. The NGTX carcinogens triggered no characteristic cancer-relevant gene profiles in all liver-based in vitro systems. In conclusion, liver-based in vitro models coupled with transcriptomics techniques, especially in the case when the HepaRG cell line is used, represent valuable tools for obtaining insight into the mechanism of action and identification of GTX carcinogens.


Subject(s)
Carcinogens/toxicity , Hepatocytes/drug effects , Liver/drug effects , Mutagens/toxicity , Transcriptome/drug effects , Animals , Carcinogens/pharmacology , Cell Line, Tumor , Embryonic Stem Cells/drug effects , Gene Expression/drug effects , Gene Expression Profiling , Hep G2 Cells , Humans , Liver Neoplasms , Mutagens/pharmacology , Rats , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/drug effects
12.
Toxicol Sci ; 124(2): 278-90, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21873647

ABSTRACT

Hepatocyte-like cells derived from the differentiation of human embryonic stem cells (hES-Hep) have potential to provide a human relevant in vitro test system in which to evaluate the carcinogenic hazard of chemicals. In this study, we have investigated this potential using a panel of 15 chemicals classified as noncarcinogens, genotoxic carcinogens, and nongenotoxic carcinogens and measured whole-genome transcriptome responses with gene expression microarrays. We applied an ANOVA model that identified 592 genes highly discriminative for the panel of chemicals. Supervised classification with these genes achieved a cross-validation accuracy of > 95%. Moreover, the expression of the response genes in hES-Hep was strongly correlated with that in human primary hepatocytes cultured in vitro. In order to infer mechanistic information on the consequences of chemical exposure in hES-Hep, we developed a computational method that measures the responses of biochemical pathways to the panel of treatments and showed that these responses were discriminative for the three toxicity classes and linked to carcinogenesis through p53, mitogen-activated protein kinases, and apoptosis pathway modules. It could further be shown that the discrimination of toxicity classes was improved when analyzing the microarray data at the pathway level. In summary, our results demonstrate, for the first time, the potential of human embryonic stem cell--derived hepatic cells as an in vitro model for hazard assessment of chemical carcinogenesis, although it should be noted that more compounds are needed to test the robustness of the assay.


Subject(s)
Carcinogenicity Tests/methods , Carcinogens/toxicity , Embryonic Stem Cells/cytology , Gene Expression Profiling , Hazardous Substances/toxicity , Hepatocytes/drug effects , Analysis of Variance , Cell Culture Techniques , Cell Differentiation , Computational Biology , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Gene Expression/drug effects , Hepatocytes/cytology , Hepatocytes/enzymology , Humans , Immunohistochemistry , Microarray Analysis , Microscopy, Phase-Contrast , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
13.
J Biotechnol ; 145(3): 284-94, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19932139

ABSTRACT

Human embryonic stem cells offer a potential unlimited supply for functional hepatocytes, since they can differentiate into hepatocyte-like cells displaying a characteristic hepatic morphology and expressing various hepatic markers. These cells could be used in various applications such as studies of drug metabolism and hepatotoxicity, which however, would require a significant expression of drug metabolizing enzymes. To derive these cells we use a stepwise differentiation protocol where growth- and maturation factors are added. The first phase involves the formation of definitive endoderm. Next, these cells are treated with factors known to promote the induction and proliferation towards hepatic progenitor cell types. In the last phase the cells are terminally differentiated and maturated into functional hepatocyte-like cells. The cultures were characterized by analysis of endodermal or hepatic markers and compared to cultures derived without induction via definitive endoderm. Hepatic functions such as urea secretion, glycogen storage, indocyanine green uptake and secretion, and cytochrome P450-expression and activity were evaluated. The DE-Hep showed a hepatocyte morphology with sub-organized cells and exhibited many liver-functions including transporter activity and capacity to metabolize drugs specific for important cytochrome P450 sub-families. This represents an important step in differentiation of hESC into functional hepatocytes.


Subject(s)
Embryonic Stem Cells/cytology , Endoderm/cytology , Hepatocytes/cytology , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Cell Line , Cell Separation , Cell Shape/drug effects , Diclofenac/metabolism , Embryonic Stem Cells/drug effects , Endoderm/drug effects , Fibroblast Growth Factor 2/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Midazolam/metabolism , Phenacetin/metabolism
14.
Stem Cells Dev ; 19(7): 961-78, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19757991

ABSTRACT

Human embryonic stem cells (hESC) can differentiate into a variety of specialized cell types, and they constitute a useful model system to study embryonic development in vitro. In order to fully utilize the potential of these cells, the mechanisms that regulate the developmental processes of specific lineage differentiation need to be better defined. The aim of this study was to explore the molecular program involved in the differentiation of hESC toward definitive endoderm (DE) and further into the hepatic lineage, and to compare that with primitive endoderm (PrE) differentiation. To that end, we applied two protocols: a specific DE differentiation protocol and an intrinsic differentiation protocol that mainly mediates PrE formation. We collected hESC, hESC-derived DE, DE-derived hepatocyte-progenitors (DE-Prog), DE-derived hepatocyte-like cells (DE-Hep), and the corresponding PrE derivatives. The samples were analyzed using microarrays, and we identified sets of genes that were exclusively up-regulated in DE derivatives (compared to PrE derivatives) at discrete developmental stages. We also investigated known protein interactions among the set of up-regulated genes in DE-Hep. The results demonstrate important differences between DE and PrE differentiation on the transcriptional level. In particular, our results identify a unique molecular program, exclusively activated during development of DE and the subsequent differentiation of DE toward the hepatic lineage. We identified key genes and pathways of potential importance for future efforts to improve hepatic differentiation from hESC. These results reveal new opportunities for rational design of specific interventions with the purpose of generating enriched populations of DE derivatives, including functional hepatocytes.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/genetics , Embryonic Stem Cells/physiology , Endoderm/cytology , Hepatocytes/cytology , Transcription, Genetic , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Endoderm/physiology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genetic Markers , Hepatocytes/physiology , Humans , Oligonucleotide Array Sequence Analysis , Up-Regulation
15.
Proc Natl Acad Sci U S A ; 103(20): 7700-5, 2006 May 16.
Article in English | MEDLINE | ID: mdl-16672370

ABSTRACT

During mammalian aging, cellular proteins become increasingly damaged: for example, by carbonylation and formation of advanced glycation end products (AGEs). The means to ensure that offspring are born without such damage are unknown. Unexpectedly, we found that undifferentiated mouse ES cells contain high levels of both carbonyls and AGEs. The damaged proteins, identified as chaperones and proteins of the cytoskeleton, are the main targets for protein oxidation in aged tissues. However, the mouse ES cells rid themselves of such damage upon differentiation in vitro. This elimination of damaged proteins coincides with a considerably elevated activity of the 20S proteasome. Moreover, damaged proteins were primarily observed in the inner cell mass of blastocysts, whereas the cells that had embarked on differentiation into the trophectoderm displayed drastically reduced levels of protein damage. Thus, the elimination of protein damage occurs also during normal embryonic development in vivo. This clear-out of damaged proteins may be a part of a previously unknown rejuvenation process at the protein level that occurs at a distinct stage during early embryonic development.


Subject(s)
Cell Differentiation/physiology , Embryo, Mammalian/physiology , Stem Cells/physiology , Aging/physiology , Animals , Cell Line , Embryo, Mammalian/anatomy & histology , Glycation End Products, Advanced/metabolism , Interleukin-6/metabolism , Leukemia Inhibitory Factor , Mice , Proteasome Endopeptidase Complex/metabolism , Stem Cells/cytology
16.
Diabetes ; 54(10): 2867-74, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16186387

ABSTRACT

The recent success in restoring normoglycemia in type 1 diabetes by islet cell transplantation indicates that cell replacement therapy of this severe disease is achievable. However, the severe lack of donor islets has increased the demand for alternative sources of beta-cells, such as adult and embryonic stem cells. Here, we investigate the potential of human embryonic stem cells (hESCs) to differentiate into beta-cells. Spontaneous differentiation of hESCs under two-dimensional growth conditions resulted in differentiation of Pdx1(+)/Foxa2(+) pancreatic progenitors and Pdx1(+)/Isl1(+) endocrine progenitors but no insulin-producing cells. However, cotransplantation of differentiated hESCs with the dorsal pancreas, but not with the liver or telencephalon, from mouse embryos resulted in differentiation of beta-cell-like cell clusters. Comparative analysis of the basic characteristics of hESC-derived insulin(+) cell clusters with human adult islets demonstrated that the insulin(+) cells share important features with normal beta-cells, such as synthesis (proinsulin) and processing (C-peptide) of insulin and nuclear localization of key beta-cell transcription factors, including Foxa2, Pdx1, and Isl1.


Subject(s)
Cell Differentiation , Embryo, Mammalian/cytology , Insulin/biosynthesis , Islets of Langerhans/cytology , Pancreas/embryology , Stem Cells/cytology , Animals , C-Peptide/analysis , C-Peptide/blood , Fluorescent Antibody Technique , Green Fluorescent Proteins/genetics , Homeodomain Proteins/analysis , Humans , Immunohistochemistry , Insulin/analysis , Islets of Langerhans/chemistry , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Pancreas/metabolism , Signal Transduction/physiology , Stem Cell Transplantation , Stem Cells/chemistry , Trans-Activators/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...