1
|
Loerch C, Szepanowski LP, Reiss J, Adjaye J, Graffmann N. Forskolin induces FXR expression and enhances maturation of iPSC-derived hepatocyte-like cells. Front Cell Dev Biol 2024; 12:1383928. [PMID: 38694820 PMCID: PMC11061433 DOI: 10.3389/fcell.2024.1383928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/04/2024] [Indexed: 05/04/2024] Open
Abstract
The generation of iPSC-derived hepatocyte-like cells (HLCs) is a powerful tool for studying liver diseases, their therapy as well as drug development. iPSC-derived disease models benefit from their diverse origin of patients, enabling the study of disease-associated mutations and, when considering more than one iPSC line to reflect a more diverse genetic background compared to immortalized cell lines. Unfortunately, the use of iPSC-derived HLCs is limited due to their lack of maturity and a rather fetal phenotype. Commercial kits and complicated 3D-protocols are cost- and time-intensive and hardly useable for smaller working groups. In this study, we optimized our previously published protocol by fine-tuning the initial cell number, exchanging antibiotics and basal medium composition and introducing the small molecule forskolin during the HLC maturation step. We thereby contribute to the liver research field by providing a simple, cost- and time-effective 2D differentiation protocol. We generate functional HLCs with significantly increased HLC hallmark gene (ALB, HNF4α, and CYP3A4) and protein (ALB) expression, as well as significantly elevated inducible CYP3A4 activity.
Collapse
Affiliation(s)
- Christiane Loerch
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Leon-Phillip Szepanowski
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- IUF – Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Julian Reiss
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- University College London, EGA Institute for Women`s Health- Zayed Center for Research Into Rare Diseases in Children (ZGR), London, United Kingdom
| | - Nina Graffmann
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
2
|
Horiuchi S, Kuroda Y, Komizu Y, Ishida S. Consideration of Commercially Available Hepatocytes as Cell Sources for Liver-Microphysiological Systems by Comparing Liver Characteristics. Pharmaceutics 2022; 15:pharmaceutics15010055. [PMID: 36678684 PMCID: PMC9867117 DOI: 10.3390/pharmaceutics15010055] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
In recent years, microphysiological systems (MPS) have been developed to shorten the test period and reduce animal experiments for drug development. We examined cell sources for the liver-MPS, i.e., MPS mimicking liver function. For liver-MPS, liver-like cells with high liver functions are required. Cryo-preserved hepatocytes (cryoheps), the gold standard hepatocytes for in vitro drug development, present several disadvantages, including differences between lots due to individual donor variations or a limited cell supply from the same donor. As such, alternatives for cryoheps are sought. Hepatocyte-like cells derived from human induced pluripotent stem cells (hiPSC-Heps), hepatocytes derived from liver-humanized mice (PXB-cells), and human liver cancer cells (HepG2 cells) were examined as source candidates for liver-MPS. Gene expression levels of the major cytochrome P450 of hiPSC-Heps, PXB cells, and HepG2 cells were compared with 22 lots of cryoheps, and the activities of hiPSC-Heps were compared with 8 lots of cryopreserved hepatocytes. A focused DNA microarray was used for the global gene analysis of the liver-like characteristics of hiPSC-Heps, PXB-cells, cryoheps, and HepG2 cells. Gene expression data from the focused microarray were analyzed by principal component analysis, hierarchical clustering, and enrichment analysis. The results indicated the characteristics of individual hepatocyte cell source and raised their consideration points as an alternative cell source candidate for liver-MPS. The study contributes to the repetitive utilization of a robust in vitro hepatic assay system over long periods with stable functionality.
Collapse
Affiliation(s)
- Shinichiro Horiuchi
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki 210-9501, Japan
| | - Yukie Kuroda
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki 210-9501, Japan
| | - Yuji Komizu
- Division of Applied Life Science, Graduate School of Engineering, Sojo University, Kumamoto 860-0082, Japan
| | - Seiichi Ishida
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki 210-9501, Japan
- Division of Applied Life Science, Graduate School of Engineering, Sojo University, Kumamoto 860-0082, Japan
- Correspondence: ; Tel.: +81-96-326-3696
| |
Collapse
|
3
|
Moreau M, Capallere C, Chavatte L, Plaza C, Meyrignac C, Pays K, Bavouzet B, Botto JM, Nizard C, Bulteau AL. Reconstruction of functional human epidermis equivalent containing 5%IPS-derived keratinocytes treated with mitochondrial stimulating plant extracts. Sci Rep 2022; 12:9073. [PMID: 35641783 PMCID: PMC9156774 DOI: 10.1038/s41598-022-13191-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 05/10/2022] [Indexed: 11/09/2022] Open
Abstract
Reconstructed human epidermis equivalents (RHE) have been developed as a clinical skin substitute and as the replacement for animal testing in both research and industry. KiPS, or keratinocytes derived from induced pluripotent stem cells (iPSCs) are frequently used to generate RHE. In this study, we focus on the mitochondrial performance of the KiPS derived from iPSCs obtained from two donors. We found that the KiPS derived from the older donor have more defective mitochondria. Treatment of these KiPS with a plant extract enriched in compounds known to protect mitochondria improved mitochondrial respiration and rendered them fully competent to derive high-quality RHE. Overall, our results suggest that improving mitochondrial function in KiPS is one of the key aspects to obtain a functional RHE and that our plant extracts can improve in this process.
Collapse
Affiliation(s)
- Marielle Moreau
- LVMH Recherche. Life Science Department, 185 Avenue de Verdun, 45800, Saint Jean de Braye, France
| | - Christophe Capallere
- Advanced Skin Research & Bioengineering Department, Ashland, Global Skin Research Center, Sophia Antipolis, France
| | - Laurent Chavatte
- Centre International de Recherche en Infectiologie, CIRI, 69007, Lyon, France.,Institut National de La Santé Et de La Recherche Médicale (INSERM) Unité U1111, 69007, Lyon, France.,Ecole Normale Supérieure de Lyon, 69007, Lyon, France.,Université Claude Bernard Lyon 1 (UCBL1), 69622, Lyon, France.,Unité Mixte de Recherche 5308 (UMR5308), Centre National de La Recherche Scientifique (CNRS), 69007, Lyon, France
| | - Christelle Plaza
- Advanced Skin Research & Bioengineering Department, Ashland, Global Skin Research Center, Sophia Antipolis, France
| | - Céline Meyrignac
- Advanced Skin Research & Bioengineering Department, Ashland, Global Skin Research Center, Sophia Antipolis, France
| | - Karl Pays
- LVMH Recherche. Life Science Department, 185 Avenue de Verdun, 45800, Saint Jean de Braye, France
| | - Bruno Bavouzet
- LVMH Recherche. Life Science Department, 185 Avenue de Verdun, 45800, Saint Jean de Braye, France
| | - Jean-Marie Botto
- Advanced Skin Research & Bioengineering Department, Ashland, Global Skin Research Center, Sophia Antipolis, France
| | - Carine Nizard
- LVMH Recherche. Life Science Department, 185 Avenue de Verdun, 45800, Saint Jean de Braye, France
| | - Anne-Laure Bulteau
- LVMH Recherche. Life Science Department, 185 Avenue de Verdun, 45800, Saint Jean de Braye, France.
| |
Collapse
|
4
|
Messina A, Luce E, Benzoubir N, Pasqua M, Pereira U, Humbert L, Eguether T, Rainteau D, Duclos-Vallée JC, Legallais C, Dubart-Kupperschmitt A. Evidence of Adult Features and Functions of Hepatocytes Differentiated from Human Induced Pluripotent Stem Cells and Self-Organized as Organoids. Cells 2022; 11:cells11030537. [PMID: 35159346 PMCID: PMC8834365 DOI: 10.3390/cells11030537] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Human-induced pluripotent stem cell-derived hepatocytes (iHeps) have been shown to have considerable potential in liver diseases, toxicity, and pharmacological studies. However, there is a growing need to obtain iHeps that are truly similar to primary adult hepatocytes in terms of morphological features and functions. We generated such human iHeps, self-assembled as organoids (iHep-Orgs). Methods: iPSC-derived hepatoblasts were self-assembled into spheroids and differentiated into mature hepatocytes modulating final step of differentiation. Results: In about four weeks of culture, the albumin secretion levels and the complete disappearance of α-fetoprotein from iHep-Orgs suggested the acquisition of a greater degree of maturation than those previously reported. The expression of apical transporters and bile acid secretion evidenced the acquisition of complex hepatocyte polarity as well as the development of a functional and well-defined bile canalicular network confirmed by computational analysis. Activities recorded for CYP450, UGT1A1, and alcohol dehydrogenase, response to hormonal stimulation, and glucose metabolism were also remarkable. Finally, iHep-Orgs displayed a considerable ability to detoxify pathological concentrations of lactate and ammonia. Conclusions: With features similar to those of primary adult hepatocytes, the iHep-Orgs thus produced could be considered as a valuable tool for the development and optimization of preclinical and clinical applications.
Collapse
Affiliation(s)
- Antonietta Messina
- UMR_S 1193, INSERM/Université Paris-Saclay, F-94800 Villejuif, France; (E.L.); (N.B.); (J.-C.D.-V.)
- Centre Hépatobiliaire, Fédération Hospitalo-Universitaire (FHU) Hépatinov, AP-HP, Hôpital Paul Brousse, F-94800 Villejuif, France; (M.P.); (U.P.); (C.L.)
- Correspondence: (A.M.); (A.D.-K.)
| | - Eléanor Luce
- UMR_S 1193, INSERM/Université Paris-Saclay, F-94800 Villejuif, France; (E.L.); (N.B.); (J.-C.D.-V.)
- Centre Hépatobiliaire, Fédération Hospitalo-Universitaire (FHU) Hépatinov, AP-HP, Hôpital Paul Brousse, F-94800 Villejuif, France; (M.P.); (U.P.); (C.L.)
| | - Nassima Benzoubir
- UMR_S 1193, INSERM/Université Paris-Saclay, F-94800 Villejuif, France; (E.L.); (N.B.); (J.-C.D.-V.)
- Centre Hépatobiliaire, Fédération Hospitalo-Universitaire (FHU) Hépatinov, AP-HP, Hôpital Paul Brousse, F-94800 Villejuif, France; (M.P.); (U.P.); (C.L.)
| | - Mattia Pasqua
- Centre Hépatobiliaire, Fédération Hospitalo-Universitaire (FHU) Hépatinov, AP-HP, Hôpital Paul Brousse, F-94800 Villejuif, France; (M.P.); (U.P.); (C.L.)
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Sorbonne Universités, 60203 Compiegne, France
| | - Ulysse Pereira
- Centre Hépatobiliaire, Fédération Hospitalo-Universitaire (FHU) Hépatinov, AP-HP, Hôpital Paul Brousse, F-94800 Villejuif, France; (M.P.); (U.P.); (C.L.)
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Sorbonne Universités, 60203 Compiegne, France
| | - Lydie Humbert
- Centre de Recherche Saint-Antoine, Sorbonne Université, INSERM, CRSA, AP-HP, Hôpital Saint Antoine, Metomics, 75012 Paris, France; (L.H.); (T.E.); (D.R.)
| | - Thibaut Eguether
- Centre de Recherche Saint-Antoine, Sorbonne Université, INSERM, CRSA, AP-HP, Hôpital Saint Antoine, Metomics, 75012 Paris, France; (L.H.); (T.E.); (D.R.)
| | - Dominique Rainteau
- Centre de Recherche Saint-Antoine, Sorbonne Université, INSERM, CRSA, AP-HP, Hôpital Saint Antoine, Metomics, 75012 Paris, France; (L.H.); (T.E.); (D.R.)
| | - Jean-Charles Duclos-Vallée
- UMR_S 1193, INSERM/Université Paris-Saclay, F-94800 Villejuif, France; (E.L.); (N.B.); (J.-C.D.-V.)
- Centre Hépatobiliaire, Fédération Hospitalo-Universitaire (FHU) Hépatinov, AP-HP, Hôpital Paul Brousse, F-94800 Villejuif, France; (M.P.); (U.P.); (C.L.)
| | - Cécile Legallais
- Centre Hépatobiliaire, Fédération Hospitalo-Universitaire (FHU) Hépatinov, AP-HP, Hôpital Paul Brousse, F-94800 Villejuif, France; (M.P.); (U.P.); (C.L.)
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Sorbonne Universités, 60203 Compiegne, France
| | - Anne Dubart-Kupperschmitt
- UMR_S 1193, INSERM/Université Paris-Saclay, F-94800 Villejuif, France; (E.L.); (N.B.); (J.-C.D.-V.)
- Centre Hépatobiliaire, Fédération Hospitalo-Universitaire (FHU) Hépatinov, AP-HP, Hôpital Paul Brousse, F-94800 Villejuif, France; (M.P.); (U.P.); (C.L.)
- Correspondence: (A.M.); (A.D.-K.)
| |
Collapse
|
5
|
Chemically Defined Conditions Mediate an Efficient Induction of Dental Pulp Pluripotent-Like Stem Cells into Hepatocyte-Like Cells. Stem Cells Int 2021; 2021:5212852. [PMID: 34795766 PMCID: PMC8593589 DOI: 10.1155/2021/5212852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 01/01/2023] Open
Abstract
Liver diseases are major causes of morbidity and mortality. Dental pulp pluripotent-like stem cells (DPPSCs) are of a considerable promise in tissue engineering and regenerative medicine as a new source of tissue-specific cells; therefore, this study is aimed at demonstrating their ability to generate functional hepatocyte-like cells in vitro. Cells were differentiated on a collagen scaffold in serum-free media supplemented with growth factors and cytokines to recapitulate liver development. At day 5, the differentiated DPPSC cells expressed the endodermal markers FOXA1 and FOXA2. Then, the cells were derived into the hepatic lineage generating hepatocyte-like cells. In addition to the associated morphological changes, the cells expressed the hepatic genes HNF6 and AFP. The terminally differentiated hepatocyte-like cells expressed the liver functional proteins albumin and CYP3A4. In this study, we report an efficient serum-free protocol to differentiate DPPSCs into functional hepatocyte-like cells. Our approach promotes the use of DPPSCs as a new source of adult stem cells for prospective use in liver regenerative medicine.
Collapse
|
6
|
Lee G, Kim H, Park JY, Kim G, Han J, Chung S, Yang JH, Jeon JS, Woo DH, Han C, Kim SK, Park HJ, Kim JH. Generation of uniform liver spheroids from human pluripotent stem cells for imaging-based drug toxicity analysis. Biomaterials 2020; 269:120529. [PMID: 33257114 DOI: 10.1016/j.biomaterials.2020.120529] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022]
Abstract
Recent advances in pluripotent stem cell technology provide an alternative source of human hepatocytes to overcome the limitations of current toxicity tests. However, this approach requires optimization and standardization before it can be used as a fast and reliable toxicity screening system. Here, we designed and tested microwell culture platforms with various diameters. We found that large quantities of uniformly-sized hepatocyte-like cell (HLC) spheroids (3D-uniHLC-Ss) could be efficiently and reproducibly generated in a short period time from a small number of differentiating human pluripotent stem cells (hPSCs). The hPSC-3D-uniHLC-Ss that were produced in 500-μm diameter microwells consistently exhibited high expressions of hepatic marker genes and had no significant signs of cell death. Importantly, a hepatic master gene hepatocyte nuclear factor 4α (HNF4α) was maintained at high levels, and the epithelial-mesenchymal transition was significantly attenuated in hPSC-3D-uniHLC-Ss. Additionally, when compared with 3D-HLC-Ss that were produced in other 3D platforms, hPSC-3D-uniHLC-Ss showed significantly higher hepatic gene expressions and drug-metabolizing activity of the enzyme, CYP3A4. Imaging-based drug toxicity studies demonstrated that hPSC-3D-uniHLC-Ss exhibited enhanced sensitivity to various hepatotoxicants, compared to HLCs, which were differentiated under 2D conditions. Precise prediction of drug-induced hepatotoxicity is a crucial step in the early phases of drug discovery. Thus, the hPSC-3D-uniHLC-Ss produced using our microwell platform could be used as an imaging-based toxicity screening system to predict drug hepatotoxicity.
Collapse
Affiliation(s)
- Gyunggyu Lee
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Hyemin Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, South Korea
| | - Ji Young Park
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Gyeongmin Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Jiyou Han
- Department of Biological Sciences, Hyupsung University, Hwasung-si, 18330, South Korea
| | - Seok Chung
- School of Mechanical Engineering, Korea University, Seoul, 20841, South Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Ji Hun Yang
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Jang Su Jeon
- Chungnam National University, Daejeon, 34134, South Korea
| | - Dong-Hun Woo
- Laboratory of Stem Cells, NEXEL Co., Ltd., Seoul, 02580, South Korea
| | - Choongseong Han
- Laboratory of Stem Cells, NEXEL Co., Ltd., Seoul, 02580, South Korea
| | - Sang Kyum Kim
- Chungnam National University, Daejeon, 34134, South Korea.
| | - Han-Jin Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, South Korea.
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|
7
|
Chaturantabut S, Shwartz A, Garnaas MK, LaBella K, Li CC, Carroll KJ, Cutting CC, Budrow N, Palaria A, Gorelick DA, Tremblay KD, North TE, Goessling W. Estrogen Acts Through Estrogen Receptor 2b to Regulate Hepatobiliary Fate During Vertebrate Development. Hepatology 2020; 72:1786-1799. [PMID: 32060934 PMCID: PMC8290048 DOI: 10.1002/hep.31184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 01/22/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS During liver development, bipotent progenitor cells differentiate into hepatocytes and biliary epithelial cells to ensure a functional liver required to maintain organismal homeostasis. The developmental cues controlling the differentiation of committed progenitors into these cell types, however, are incompletely understood. Here, we discover an essential role for estrogenic regulation in vertebrate liver development to affect hepatobiliary fate decisions. APPROACH AND RESULTS Exposure of zebrafish embryos to 17β-estradiol (E2) during liver development significantly decreased hepatocyte-specific gene expression, liver size, and hepatocyte number. In contrast, pharmacological blockade of estrogen synthesis or nuclear estrogen receptor (ESR) signaling enhanced liver size and hepatocyte marker expression. Transgenic reporter fish demonstrated nuclear ESR activity in the developing liver. Chemical inhibition and morpholino knockdown of nuclear estrogen receptor 2b (esr2b) increased hepatocyte gene expression and blocked the effects of E2 exposure. esr2b-/- mutant zebrafish exhibited significantly increased expression of hepatocyte markers with no impact on liver progenitors, other endodermal lineages, or vasculature. Significantly, E2-stimulated Esr2b activity promoted biliary epithelial differentiation at the expense of hepatocyte fate, whereas loss of esr2b impaired biliary lineage commitment. Chemical and genetic epistasis studies identified bone morphogenetic protein (BMP) signaling as a mediator of the estrogen effects. The divergent impact of estrogen on hepatobiliary fate was confirmed in a human hepatoblast cell line, indicating the relevance of this pathway for human liver development. CONCLUSIONS Our studies identify E2, esr2b, and downstream BMP activity as important regulators of hepatobiliary fate decisions during vertebrate liver development. These results have significant clinical implications for liver development in infants exposed to abnormal estrogen levels or estrogenic compounds during pregnancy.
Collapse
Affiliation(s)
| | - Arkadi Shwartz
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Maija K. Garnaas
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kyle LaBella
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Chia-Cheng Li
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kelli J. Carroll
- Stem Cell Program, Division of Hematology-Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Claire C. Cutting
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Nadine Budrow
- Stem Cell Program, Division of Hematology-Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Amrita Palaria
- Department of Animal and Veterinary Sciences, University of Massachusetts, Amherst, MA, USA
| | - Daniel A. Gorelick
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Kimberly D. Tremblay
- Department of Animal and Veterinary Sciences, University of Massachusetts, Amherst, MA, USA
| | - Trista E. North
- Stem Cell Program, Division of Hematology-Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Wolfram Goessling
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA.,Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
8
|
Hausmann C, Vogt A, Kerscher M, Ghoreschi K, Schäfer-Korting M, Zoschke C. Optimizing skin pharmacotherapy for older patients: the future is at hand but are we ready for it? Drug Discov Today 2020; 25:851-861. [PMID: 31987937 DOI: 10.1016/j.drudis.2020.01.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/04/2020] [Accepted: 01/16/2020] [Indexed: 02/06/2023]
Abstract
Age-related changes affect both the local pharmacotherapy of skin diseases and the transdermal administration of drugs. The development of aged skin models disregards the highly individual process of aging, facilitating general conclusions for older patients. Nevertheless, 'omics technology, high-content screening, and non-invasive imaging, as well as bioprinting, CRISPR-Cas, and, patients-on-a-chip, can retrieve personalized information for the generation of in vitro models. Herein, we suggest a strategy to optimize pharmacotherapy for older patients. The technology for relevant human cell-based models is at hand and the consideration of patient heterogeneity is required to unlock their full potential.
Collapse
Affiliation(s)
- Christian Hausmann
- Freie Universität Berlin, Institute of Pharmacy (Pharmacology & Toxicology), Königin-Luise-Strasse 2+4, 14195 Berlin, Germany
| | - Annika Vogt
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Dermatology, Venereology and Allergology, Charitéplatz 1, 10117 Berlin, Germany
| | - Martina Kerscher
- Universität Hamburg, Division of Biochemistry and Molecular Biology, Papendamm 21, 20146 Hamburg, Germany
| | - Kamran Ghoreschi
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Dermatology, Venereology and Allergology, Charitéplatz 1, 10117 Berlin, Germany
| | - Monika Schäfer-Korting
- Freie Universität Berlin, Institute of Pharmacy (Pharmacology & Toxicology), Königin-Luise-Strasse 2+4, 14195 Berlin, Germany
| | - Christian Zoschke
- Freie Universität Berlin, Institute of Pharmacy (Pharmacology & Toxicology), Königin-Luise-Strasse 2+4, 14195 Berlin, Germany.
| |
Collapse
|
9
|
Guan S, Zhang K, Li J. Recent Advances in Extracellular Matrix for Engineering Stem Cell Responses. Curr Med Chem 2019; 26:6321-6338. [DOI: 10.2174/0929867326666190704121309] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/02/2018] [Accepted: 01/25/2019] [Indexed: 02/06/2023]
Abstract
Stem cell transplantation is an advanced medical technology, which brings hope for the
treatment of some difficult diseases in the clinic. Attributed to its self-renewal and differential
ability, stem cell research has been pushed to the forefront of regenerative medicine and has become
a hot topic in tissue engineering. The surrounding extracellular matrix has physical functions
and important biological significance in regulating the life activities of cells, which may play crucial
roles for in situ inducing specific differentiation of stem cells. In this review, we discuss the
stem cells and their engineering application, and highlight the control of the fate of stem cells, we
offer our perspectives on the various challenges and opportunities facing the use of the components
of extracellular matrix for stem cell attachment, growth, proliferation, migration and differentiation.
Collapse
Affiliation(s)
- Shuaimeng Guan
- School of Life Science, Zhengzhou University, Zhengzhou 450000, China
| | - Kun Zhang
- School of Life Science, Zhengzhou University, Zhengzhou 450000, China
| | - Jingan Li
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450000, China
| |
Collapse
|
10
|
Advances in Human Induced Pluripotent Stem Cell-Derived Hepatocytes for Use in Toxicity Testing. Ann Biomed Eng 2019; 48:1045-1057. [PMID: 31372857 DOI: 10.1007/s10439-019-02331-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 07/19/2019] [Indexed: 02/08/2023]
Abstract
Induced pluripotent stem cells (iPSCs) can be differentiated into multiple cell types in the body while maintaining proliferative capabilities. The generation of hepatocyte-like cells (HLCs) from iPSCs has resulted in a new source for liver cells. Since healthy primary human hepatocytes and hepatic cells are difficult to obtain, HLCs are gaining attention. HLCs can be obtained from a continuous, stable source while maintaining their original donor genotype, which opens new avenues into patient-specific testing and therapeutics. Studies have utilized HLCs for toxicity testing to further understand their drug metabolizing capabilities. This review focuses on advances being made to achieve hepatic functions from HLCs, their current use in hepatotoxicity testing, and their potential for future liver-related toxicity evaluations.
Collapse
|
11
|
Wnorowski A, Yang H, Wu JC. Progress, obstacles, and limitations in the use of stem cells in organ-on-a-chip models. Adv Drug Deliv Rev 2019; 140:3-11. [PMID: 29885330 DOI: 10.1016/j.addr.2018.06.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/16/2018] [Accepted: 06/01/2018] [Indexed: 12/18/2022]
Abstract
In recent years, drug development costs have soared, primarily due to the failure of preclinical animal and cell culture models, which do not directly translate to human physiology. Organ-on-a-chip (OOC) is a burgeoning technology with the potential to revolutionize disease modeling, drug discovery, and toxicology research by strengthening the relevance of culture-based models while reducing costly animal studies. Although OOC models can incorporate a variety of tissue sources, the most robust and relevant OOC models going forward will include stem cells. In this review, we will highlight the benefits of stem cells as a tissue source while considering current limitations to their complete and effective implementation into OOC models.
Collapse
Affiliation(s)
- Alexa Wnorowski
- Stanford Cardiovascular Institute, Stanford, CA 94305, United States; Department of Bioengineering, Stanford University Schools of Engineering and Medicine, Stanford, CA 943055, United States
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford, CA 94305, United States
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford, CA 94305, United States; Division of Cardiovascular Medicine, Department of Medicine, Stanford, CA 94305, United States; Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, United States.
| |
Collapse
|
12
|
Lereau Bernier M, Poulain S, Tauran Y, Danoy M, Shinohara M, Kimura K, Segard BD, Kato S, Kido T, Miyajima A, Sakai Y, Plessy C, Leclerc É. Profiling of derived-hepatocyte progenitors from induced pluripotent stem cells using nanoCAGE promoter analysis. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2018.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
13
|
Wang YM, Li K, Dou XG, Bai H, Zhao XP, Ma X, Li LJ, Chen ZS, Huang YC. Treatment of AECHB and Severe Hepatitis (Liver Failure). ACUTE EXACERBATION OF CHRONIC HEPATITIS B 2019. [PMCID: PMC7498915 DOI: 10.1007/978-94-024-1603-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This chapter describes the general treatment and immune principles and internal management for AECHB and HBV ACLF, including ICU monitoring, general supportive medications/nutrition/nursing, immune therapy, artificial liver supportive systems, hepatocyte/stem cell, and liver transplant, management for special populations, frequently clinical complications and the utilization of Chinese traditional medicines.Early clinical indicators of severe hepatitis B include acratia, gastrointestinal symptoms, a daily increase in serum bilirubin >1 mg/dL, toxic intestinal paralysis, bleeding tendency and mild mind anomaly or character change, and the presence of other diseases inducing severe hepatitis. Laboratory indicators include T-Bil, PTA, cholinesterase, pre-albumin and albumin. The roles of immune indicators (such as IL-6, TNF-α, and fgl2), gene polymorphisms, HBV genotypes, and gene mutations as early clinical indicators. Intensive Care Unit monitor patients with severe hepatitis include intracranial pressure, infection, blood dynamics, respiratory function, renal function, blood coagulation function, nutritional status and blood purification process. Nursing care should not only include routine care, but psychological and special care (complications). Nutrition support and nursing care should be maintained throughout treatment for severe hepatitis. Common methods of evaluating nutritional status include direct human body measurement, creatinine height index (CHI) and subject global assessment of nutrition (SGA). Malnourished patients should receive enteral or parenteral nutrition support. Immune therapies for severe hepatitis include promoting hepatocyte regeneration (e.g. with glucagon, hepatocyte growth factor and prostaglandin E1), glucocorticoid suppressive therapy, and targeting molecular blocking. Corticosteroid treatment should be early and sufficient, and adverse drug reactions monitored. Treatments currently being investigated are those targeting Toll-like receptors, NK cell/NK cell receptors, macrophage/immune coagulation system, CTLA-4/PD-1 and stem cell transplantation. In addition to conventional drugs and radioiodine, corticosteroids and artificial liver treatment can also be considered for severe hepatitis patients with hyperthyreosis. Patients with gestational severe hepatitis require preventive therapy for fetal growth restriction, and it is necessary to choose the timing and method of fetal delivery. For patients with both diabetes and severe hepatitis, insulin is preferred to oral antidiabetic agents to control blood glucose concentration. Liver toxicity of corticosteroids and immune suppressors should be monitored during treatment for severe hepatitis in patients with connective tissue diseases including SLE, RA and sicca syndrome. Patient with connective tissue diseases should preferably be started after the antiviral treatment with nucleos(t)ide analogues. An artificial liver can improve patients’ liver function; remove endotoxins, blood ammonia and other toxins; correct amino acid metabolism and coagulation disorders; and reverse internal environment imbalances. Non-bioartificial livers are suitable for patients with early and middle stage severe hepatitis; for late-stage patients waiting for liver transplantation; and for transplanted patients with rejection reaction or transplant failure. The type of artificial liver should be determined by each patient’s condition and previous treatment purpose, and patients should be closely monitored for adverse reactions and complications. Bio- and hybrid artificial livers are still under development. MELD score is the international standard for choosing liver transplantation. Surgical methods mainly include the in situ classic type and the piggyback type; transplantation includes no liver prophase, no liver phase or new liver phase. Preoperative preparation, management of intraoperative and postoperative complications and postoperative long-term treatment are keys to success. Severe hepatitis belongs to the categories of “acute jaundice”, “scourge jaundice”, and “hot liver” in traditional Chinese medicine. Treatment methods include Chinese traditional medicines, acupuncture and acupoint injection, external application of drugs, umbilical compress therapy, drip, blow nose therapy, earpins, and clysis. Dietary care is also an important part of traditional Chinese medicine treatment.
Collapse
|
14
|
Analysis of differentially expressed genes among human hair follicle-derived iPSCs, induced hepatocyte-like cells, and primary hepatocytes. Stem Cell Res Ther 2018; 9:211. [PMID: 30092828 PMCID: PMC6085644 DOI: 10.1186/s13287-018-0940-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/07/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022] Open
Abstract
Background Differentiation of human induced pluripotent stem cells (hiPSCs) into hepatocytes has important clinical significance in providing a new stem cell source for cell therapy of terminal liver disease. The differential gene expression analysis of hiPSCs, induced hepatocyte-like cells (HLCs), and primary human hepatocytes (PHHs) provides valuable information for optimization of an induction scheme and exploration of differentiation mechanisms. Methods Human hair follicle-derived iPSCs (hHF-iPSCs) were induced in vitro by mimicking the environment of a developing liver for 19 days. Expression of specific proteins was determined by immunofluorescence staining; the function of HLCs in storage and metabolism was identified by detecting periodic acid–Schiff, indocyanine green, and low-density lipoprotein. Based on the transcriptomics data, the differential gene expression profiles of hHF-iPSCs, HLCs, and PHHs were analyzed by Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway, FunRich, and network analysis methods. Results HLCs were able to express albumin (ALB), alpha-fetoprotein, CYP3A4, and CYP7A1, and exhibited matured liver cell functions such as glycogen synthesis and storage. Complement and coagulation cascades and metabolic pathways ranked top in the downregulated list of HLCs/PHHs, while the cell cycle ranked top in the upregulated list of HLCs/PHHs. In the protein–protein interaction network, according to the degree rankings, TOP2A, CDK1, etc. were the important upregulated differentially expressed genes (DEGs), while ALB, ACACB, etc. were the major downregulated DEGs in HLCs/PHHs; the module analysis indicated that CDCA8, AURKB, and AURKA were the top upregulated DEGs in HLCs/PHHs. Conclusions We presented the differences in gene expression among hHF-iPSCs, HLCs, and PHHs through transcriptome array data and provided new ideas for the optimization of induction. Electronic supplementary material The online version of this article (10.1186/s13287-018-0940-z) contains supplementary material, which is available to authorized users.
Collapse
|
15
|
Li L, Miu KK, Gu S, Cheung HH, Chan WY. Comparison of multi-lineage differentiation of hiPSCs reveals novel miRNAs that regulate lineage specification. Sci Rep 2018; 8:9630. [PMID: 29941943 PMCID: PMC6018499 DOI: 10.1038/s41598-018-27719-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 06/07/2018] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are known to be crucial players in governing the differentiation of human induced pluripotent stem cells (hiPSCs). Despite their utter importance, identifying key lineage specifiers among the myriads of expressed miRNAs remains challenging. We believe that the current practice in mining miRNA specifiers via delineating dynamic fold-changes only is inadequate. Our study, therefore, provides evidence to pronounce "lineage specificity" as another important attribute to qualify for these lineage specifiers. Adopted hiPSCs were differentiated into representative lineages (hepatic, nephric and neuronal) over all three germ layers whilst the depicted miRNA expression changes compiled into an integrated atlas. We demonstrated inter-lineage analysis shall aid in the identification of key miRNAs with lineage-specificity, while these shortlisted candidates were collectively known as "lineage-specific miRNAs". Subsequently, we followed through the fold-changes along differentiation via computational analysis to identify miR-192 and miR-372-3p, respectively, as representative candidate key miRNAs for the hepatic and nephric lineages. Indeed, functional characterization validated that miR-192 and miR-372-3p regulate lineage differentiation via modulation of the expressions of lineage-specific genes. In summary, our presented miRNA atlas is a resourceful ore for the mining of key miRNAs responsible for lineage specification.
Collapse
Affiliation(s)
- Lu Li
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR
- School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Kai-Kei Miu
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR
| | - Shen Gu
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR
- M&H Genetics/Baylor Genetics Laboratories, Baylor College of Medicine, Houston, TX, USA
| | - Hoi-Hung Cheung
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR.
| | - Wai-Yee Chan
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR.
| |
Collapse
|
16
|
Raju R, Chau D, Notelaers T, Myers CL, Verfaillie CM, Hu WS. In Vitro Pluripotent Stem Cell Differentiation to Hepatocyte Ceases Further Maturation at an Equivalent Stage of E15 in Mouse Embryonic Liver Development. Stem Cells Dev 2018; 27:910-921. [PMID: 29851366 DOI: 10.1089/scd.2017.0270] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hepatocyte-like cells (HLCs) can be derived from pluripotent stem cells (PSCs) by sequential treatment of chemical cues to mimic the microenvironment of embryonic liver development. However, these HLCs do not reach the full maturity level of primary hepatocytes. In this study, we carried out a meta-analysis of cross-species transcriptome data of in vitro differentiation of human PSCs to HLCs and in vivo mouse embryonic liver development to identify the developmental stage at which HLC maturation was blocked at. Systematic variations were found associated with the data source and removed by batch correction. Using principal component analysis, HLCs from different stages of differentiation were aligned with mouse embryonic liver development chronologically. A "unified developmental time" (DT) scale was developed after aligning in vitro HLC differentiation and in vivo embryonic liver development. HLCs were found to cease further maturation at an equivalent stage of mouse embryonic day (E)13-15. Genes with discordant time dynamics were identified by aligning in vivo and in vitro data set onto a common DT scale. These genes may be targets of genetic intervention for enhancing the maturity of PSC-derived HLCs.
Collapse
Affiliation(s)
- Ravali Raju
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| | - David Chau
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota.,3 Department of Biomedical Engineering, University of Minnesota , Minneapolis, Minnesota
| | - Tineke Notelaers
- 4 Department of Development and Regeneration, KU Leuven , Leuven, Belgium .,5 Stem Cell Institute Leuven , KU Leuven, Leuven, Belgium
| | - Chad L Myers
- 6 Department of Computer Science and Engineering, University of Minnesota , Minneapolis, Minnesota
| | - Catherine M Verfaillie
- 4 Department of Development and Regeneration, KU Leuven , Leuven, Belgium .,5 Stem Cell Institute Leuven , KU Leuven, Leuven, Belgium
| | - Wei-Shou Hu
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|
17
|
Abstract
Induced pluripotent stem cells (iPSCs) and human embryonic stem cells (hESCs) differentiated into hepatocyte-like cells (HLCs) provide a defined and renewable source of cells for drug screening, toxicology and regenerative medicine. We previously reprogrammed human fetal foreskin fibroblast cells (HFF1) into iPSCs employing an episomal plasmid-based integration-free approach, this iPSC-line and the hESC lines H1 and H9 were used to model hepatogenesis in vitro. Biochemical characterisation confirmed glycogen storage, ICG uptake and release, urea and bile acid production, as well as CYP3A4 activity. Microarray-based transcriptome analyses was carried out using RNA isolated from the undifferentiated pluripotent stem cells and subsequent differentiation stages- definitive endoderm (DE) hepatic endoderm (HE) and HLCs. K-means identified 100 distinct clusters, for example, POU5F1/OCT4 marking the undifferentiated stage, SOX17 the DE stage, HNF4α the HE stage, and ALB specific to HLCs, fetal liver and primary human hepatocytes (PHH). This data descriptor describes these datasets which should be useful for gaining new insights into the molecular basis of hepatogenesis and associated gene regulatory networks.
Collapse
|
18
|
Du C, Feng Y, Qiu D, Xu Y, Pang M, Cai N, Xiang AP, Zhang Q. Highly efficient and expedited hepatic differentiation from human pluripotent stem cells by pure small-molecule cocktails. Stem Cell Res Ther 2018. [PMID: 29523187 PMCID: PMC5845228 DOI: 10.1186/s13287-018-0794-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background The advent of human-induced pluripotent stem cells holds great promise for producing ample individualized hepatocytes. Although previous efforts have succeeded in generating hepatocytes from human pluripotent stem cells in vitro by viral-based expression of transcription factors and/or addition of growth factors during the differentiation process, the safety issue of viral transduction and high cost of cytokines would hinder the downstream applications. Recently, the use of small molecules has emerged as a powerful tool to induce cell fate transition for their superior stability, safety, cell permeability, and cost-effectiveness. Methods In the present study, we established a novel efficient hepatocyte differentiation strategy of human pluripotent stem cells with pure small-molecule cocktails. This method induced hepatocyte differentiation in a stepwise manner, including definitive endoderm differentiation, hepatic specification, and hepatocyte maturation within only 13 days. Results The differentiated hepatic-like cells were morphologically similar to hepatocytes derived from growth factor-based methods and primary hepatocytes. These cells not only expressed specific hepatic markers at the transcriptional and protein levels, but also possessed main liver functions such as albumin production, glycogen storage, cytochrome P450 activity, and indocyanine green uptake and release. Conclusions Highly efficient and expedited hepatic differentiation from human pluripotent stem cells could be achieved by our present novel, pure, small-molecule cocktails strategy, which provides a cost-effective platform for in vitro studies of the molecular mechanisms of human liver development and holds significant potential for future clinical applications. Electronic supplementary material The online version of this article (10.1186/s13287-018-0794-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cong Du
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China.,Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China
| | - Yuan Feng
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China.,Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China
| | - Dongbo Qiu
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China.,Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China
| | - Yan Xu
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China.,Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China
| | - Mao Pang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China
| | - Nan Cai
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China.,Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China
| | - Andy Peng Xiang
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China.,Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Qi Zhang
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China. .,Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China. .,Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China. .,Biotherapy Center & Cell-gene Therapy Translational Medicine Research Center, Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
19
|
Maguire EM, Xiao Q, Xu Q. Differentiation and Application of Induced Pluripotent Stem Cell–Derived Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2017; 37:2026-2037. [DOI: 10.1161/atvbaha.117.309196] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/21/2017] [Indexed: 02/06/2023]
Abstract
Vascular smooth muscle cells (VSMCs) play a role in the development of vascular disease, for example, neointimal formation, arterial aneurysm, and Marfan syndrome caused by genetic mutations in VSMCs, but little is known about the mechanisms of the disease process. Advances in induced pluripotent stem cell technology have now made it possible to derive VSMCs from several different somatic cells using a selection of protocols. As such, researchers have set out to delineate key signaling processes involved in triggering VSMC gene expression to grasp the extent of gene regulatory networks involved in phenotype commitment. This technology has also paved the way for investigations into diseases affecting VSMC behavior and function, which may be treatable once an identifiable culprit molecule or gene has been repaired. Moreover, induced pluripotent stem cell–derived VSMCs are also being considered for their use in tissue-engineered blood vessels as they may prove more beneficial than using autologous vessels. Finally, while several issues remains to be clarified before induced pluripotent stem cell–derived VSMCs can become used in regenerative medicine, they do offer both clinicians and researchers hope for both treating and understanding vascular disease. In this review, we aim to update the recent progress on VSMC generation from stem cells and the underlying molecular mechanisms of VSMC differentiation. We will also explore how the use of induced pluripotent stem cell–derived VSMCs has changed the game for regenerative medicine by offering new therapeutic avenues to clinicians, as well as providing researchers with a new platform for modeling of vascular disease.
Collapse
Affiliation(s)
- Eithne Margaret Maguire
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (E.M.M., Q. Xiao); and Cardiovascular Division, King’s College London BHF Centre, United Kingdom (Q. Xu)
| | - Qingzhong Xiao
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (E.M.M., Q. Xiao); and Cardiovascular Division, King’s College London BHF Centre, United Kingdom (Q. Xu)
| | - Qingbo Xu
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (E.M.M., Q. Xiao); and Cardiovascular Division, King’s College London BHF Centre, United Kingdom (Q. Xu)
| |
Collapse
|
20
|
Footprint-free human fetal foreskin derived iPSCs: A tool for modeling hepatogenesis associated gene regulatory networks. Sci Rep 2017; 7:6294. [PMID: 28740077 PMCID: PMC5524812 DOI: 10.1038/s41598-017-06546-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 06/13/2017] [Indexed: 12/17/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are similar to embryonic stem cells and can be generated from somatic cells. We have generated episomal plasmid-based and integration-free iPSCs (E-iPSCs) from human fetal foreskin fibroblast cells (HFF1). We used an E-iPSC-line to model hepatogenesis in vitro. The HLCs were characterized biochemically, i.e. glycogen storage, ICG uptake and release, UREA and bile acid production, as well as CYP3A4 activity. Ultra-structure analysis by electron microscopy revealed the presence of lipid and glycogen storage, tight junctions and bile canaliculi- all typical features of hepatocytes. Furthermore, the transcriptome of undifferentiated E-iPSC, DE, HE and HLCs were compared to that of fetal liver and primary human hepatocytes (PHH). K-means clustering identified 100 clusters which include developmental stage-specific groups of genes, e.g. OCT4 expression at the undifferentiated stage, SOX17 marking the DE stage, DLK and HNF6 the HE stage, HNF4α and Albumin is specific to HLCs, fetal liver and adult liver (PHH) stage. We use E-iPSCs for modeling gene regulatory networks associated with human hepatogenesis and gastrulation in general.
Collapse
|
21
|
Kochat V, Equbal Z, Baligar P, Kumar V, Srivastava M, Mukhopadhyay A. JMJD3 aids in reprogramming of bone marrow progenitor cells to hepatic phenotype through epigenetic activation of hepatic transcription factors. PLoS One 2017; 12:e0173977. [PMID: 28328977 PMCID: PMC5362104 DOI: 10.1371/journal.pone.0173977] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/01/2017] [Indexed: 01/23/2023] Open
Abstract
The strictly regulated unidirectional differentiation program in some somatic stem/progenitor cells has been found to be modified in the ectopic site (tissue) undergoing regeneration. In these cases, the lineage barrier is crossed by either heterotypic cell fusion or direct differentiation. Though studies have shown the role of coordinated genetic and epigenetic mechanisms in cellular development and differentiation, how the lineage fate of adult bone marrow progenitor cells (BMPCs) is reprogrammed during liver regeneration and whether this lineage switch is stably maintained are not clearly understood. In the present study, we wanted to decipher genetic and epigenetic mechanisms that involve in lineage reprogramming of BMPCs into hepatocyte-like cells. Here we report dynamic transcriptional change during cellular reprogramming of BMPCs to hepatocytes and dissect the epigenetic switch mechanism of BM cell-mediated liver regeneration after acute injury. Genome-wide gene expression analysis in BM-derived hepatocytes, isolated after 1 month and 5 months of transplantation, showed induction of hepatic transcriptional program and diminishing of donor signatures over the time. The transcriptional reprogramming of BM-derived cells was found to be the result of enrichment of activating marks (H3K4me3 and H3K9Ac) and loss of repressive marks (H3K27me3 and H3K9me3) at the promoters of hepatic transcription factors (HTFs). Further analyses showed that BMPCs possess bivalent histone marks (H3K4me3 and H3K27me3) at the promoters of crucial HTFs. H3K27 methylation dynamics at the HTFs was antagonistically regulated by EZH2 and JMJD3. Preliminary evidence suggests a role of JMJD3 in removal of H3K27me3 mark from promoters of HTFs, thus activating epigenetically poised hepatic genes in BMPCs prior to partial nuclear reprogramming. The importance of JMJD3 in reprogramming of BMPCs to hepatic phenotype was confirmed by inhibiting catalytic function of the enzyme using small molecule GSK-J4. Our results propose a potential role of JMJD3 in lineage conversion of BM cells into hepatic lineage.
Collapse
Affiliation(s)
- Veena Kochat
- Stem Cell Biology Laboratory, National Institute of Immunology, New Delhi, India
| | - Zaffar Equbal
- Stem Cell Biology Laboratory, National Institute of Immunology, New Delhi, India
| | - Prakash Baligar
- Stem Cell Biology Laboratory, National Institute of Immunology, New Delhi, India
| | - Vikash Kumar
- Stem Cell Biology Laboratory, National Institute of Immunology, New Delhi, India
| | - Madhulika Srivastava
- Epigenetic Regulation Research Laboratory, National Institute of Immunology, New Delhi, India
| | - Asok Mukhopadhyay
- Stem Cell Biology Laboratory, National Institute of Immunology, New Delhi, India
- * E-mail: ,
| |
Collapse
|
22
|
Sistare FD, Mattes WB, LeCluyse EL. The Promise of New Technologies to Reduce, Refine, or Replace Animal Use while Reducing Risks of Drug Induced Liver Injury in Pharmaceutical Development. ILAR J 2017; 57:186-211. [DOI: 10.1093/ilar/ilw025] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 07/25/2016] [Accepted: 09/13/2016] [Indexed: 12/19/2022] Open
|
23
|
Graffmann N, Ring S, Kawala MA, Wruck W, Ncube A, Trompeter HI, Adjaye J. Modeling Nonalcoholic Fatty Liver Disease with Human Pluripotent Stem Cell-Derived Immature Hepatocyte-Like Cells Reveals Activation of PLIN2 and Confirms Regulatory Functions of Peroxisome Proliferator-Activated Receptor Alpha. Stem Cells Dev 2016; 25:1119-33. [PMID: 27308945 PMCID: PMC4971413 DOI: 10.1089/scd.2015.0383] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD/steatosis) is a metabolic disease characterized by the incorporation of fat into hepatocytes. In this study, we developed an in vitro model for NAFLD based on hepatocyte-like cells (HLCs) differentiated from human pluripotent stem cells. We induced fat storage in these HLCs and detected major expression changes of metabolism-associated genes, as well as an overall reduction of liver-related microRNAs. We observed an upregulation of the lipid droplet coating protein Perilipin 2 (PLIN2), as well as of numerous genes of the peroxisome proliferator-activated receptor (PPAR) pathway, which constitutes a regulatory hub for metabolic processes. Interference with PLIN2 and PPARα resulted in major alterations in gene expression, especially affecting lipid, glucose, and purine metabolism. Our model recapitulates many metabolic changes that are characteristic for NAFLD. It permits the dissection of disease-promoting molecular pathways and allows us to investigate the influences of distinct genetic backgrounds on disease progression.
Collapse
Affiliation(s)
- Nina Graffmann
- 1 Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf , Düsseldorf, Germany
| | - Sarah Ring
- 1 Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf , Düsseldorf, Germany
| | - Marie-Ann Kawala
- 1 Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf , Düsseldorf, Germany
| | - Wasco Wruck
- 1 Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf , Düsseldorf, Germany
| | - Audrey Ncube
- 1 Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf , Düsseldorf, Germany
| | - Hans-Ingo Trompeter
- 2 Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Düsseldorf , Düsseldorf, Germany
| | - James Adjaye
- 1 Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf , Düsseldorf, Germany
| |
Collapse
|
24
|
Wruck W, Graffmann N, Kawala MA, Adjaye J. Concise Review: Current Status and Future Directions on Research Related to Nonalcoholic Fatty Liver Disease. Stem Cells 2016; 35:89-96. [PMID: 27374784 DOI: 10.1002/stem.2454] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/27/2016] [Accepted: 06/18/2016] [Indexed: 02/06/2023]
Abstract
Considered a feature of the metabolic syndrome, nonalcoholic fatty liver disease (NAFLD), is associated with insulin resistance, type 2 diabetes, obesity and drug toxicity. Its prevalence is estimated at about 30% in western countries mainly due to sedentary life styles and high fat diets. Genome-wide association studies have identified polymorphisms in several genes, for example, PNPLA3, and TM6SF2 which confer susceptibility to NAFLD. Here, we review recent findings in the NAFLD field with a particular focus on published transcriptomics datasets which we subject to a meta-analysis. We reveal a common gene signature correlating with the progression of the disease from steatosis and steatohepatitis and reveal that lipogenic and cholesterol metabolic pathways are main actors in this signature. We propose the use of disease-in-a-dish models based on hepatocyte-like cells derived from patient-specific induced pluripotent stem cells (iPSC). These will enable investigations into the contribution of genetic background in the progression from NALFD to non-alcoholic steatohepatitis. Furthermore, an iPSC-based approach should aid in the elucidation of the function of new biomarkers, thus enabling better diagnostic tests and validation of potential drug targets. Stem Cells 2017;35:89-96.
Collapse
Affiliation(s)
- Wasco Wruck
- Medical Faculty, Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University, Düsseldorf, Germany
| | - Nina Graffmann
- Medical Faculty, Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University, Düsseldorf, Germany
| | - Marie-Ann Kawala
- Medical Faculty, Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University, Düsseldorf, Germany
| | - James Adjaye
- Medical Faculty, Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
25
|
Wang L, Su W, Du W, Xu Y, Wang L, Kong D, Han Z, Zheng G, Li Z. Gene and MicroRNA Profiling of Human Induced Pluripotent Stem Cell-Derived Endothelial Cells. Stem Cell Rev Rep 2016; 11:219-27. [PMID: 25618294 DOI: 10.1007/s12015-014-9582-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION The differentiated cell lineages from human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) have shown their potential in regenerative medicine. However, the functional and transcriptional microRNA (miRNA) expression pattern during endothelial differentiation has yet to be characterized. METHODS In this study, hESCs and hiPSCs were differentiated into endothelial cells (ECs). Then the endothelial-related gene profiling and miRNA profiling of hiPSCs, hESCs, hiPSCs derived endothelial cells (hiPSC-ECs), hESC derived endothelial cells (hESC-ECs) and human umbilical vein endothelial cells (HUVECs) were compared using RT-PCR Array. The data was analyzed using the data analysis system on QIAGEN's website. RESULTS Our analysis demonstrated that the endothelial differentiation was triggered after EB formation and the EC-associated genes were up-regulated swiftly in both hESC-EBs and hiPSC-EBs; hiPSC-ECs and hESC-ECs had the similar EC-associated gene expression patterns. Moreover, we report here the first miRNA profiling study of hiPSC-ECs. Analyzing 376 unique miRNAs, we have identified several interesting miRNAs, including miR-20a, miR-20b, miR-222, miR-210, which have been previously reported to be involved in endothelial differentiation and show surprising expression patterns across our samples. We also identified novel miRNAs, such as miR-125a-5p, miR-149, miR-296-5p, miR-100, miR-27b, miR-181a and miR-137, which were up-regulated in both hiPSC-ECs and hESC-ECs during endothelial differentiation. CONCLUSION hiPSC-ECs and hESC-ECs exhibited a high degree of similarity with HUVECs in EC-associated genes expression. And the miRNA profiling analysis revealed significant differences between hiPSCs and hESCs, but a high degree of similarity between hiPSC-ECs and hESC-ECs.
Collapse
Affiliation(s)
- Lina Wang
- State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Stem Cells and Regenerative Medicine: Myth or Reality of the 21th Century. Stem Cells Int 2015; 2015:734731. [PMID: 26300923 PMCID: PMC4537770 DOI: 10.1155/2015/734731] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 04/22/2015] [Accepted: 05/24/2015] [Indexed: 02/07/2023] Open
Abstract
Since the 1960s and the therapeutic use of hematopoietic stem cells of bone marrow origin, there has been an increasing interest in the study of undifferentiated progenitors that have the ability to proliferate and differentiate into various tissues. Stem cells (SC) with different potency can be isolated and characterised. Despite the promise of embryonic stem cells, in many cases, adult or even fetal stem cells provide a more interesting approach for clinical applications. It is undeniable that mesenchymal stem cells (MSC) from bone marrow, adipose tissue, or Wharton's Jelly are of potential interest for clinical applications in regenerative medicine because they are easily available without ethical problems for their uses. During the last 10 years, these multipotent cells have generated considerable interest and have particularly been shown to escape to allogeneic immune response and be capable of immunomodulatory activity. These properties may be of a great interest for regenerative medicine. Different clinical applications are under study (cardiac insufficiency, atherosclerosis, stroke, bone and cartilage deterioration, diabetes, urology, liver, ophthalmology, and organ's reconstruction). This review focuses mainly on tissue and organ regeneration using SC and in particular MSC.
Collapse
|
27
|
Lu J, Einhorn S, Venkatarangan L, Miller M, Mann DA, Watkins PB, LeCluyse E. Morphological and Functional Characterization and Assessment of iPSC-Derived Hepatocytes for In Vitro Toxicity Testing. Toxicol Sci 2015; 147:39-54. [PMID: 26092927 DOI: 10.1093/toxsci/kfv117] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Drug-induced liver injury (DILI) remains a great challenge and a major concern during late-stage drug development. Induced pluripotent stem cells (iPSC) represent an exciting alternative in vitro model system to explore the role of genetic diversity in DILI, especially when derived from patients who have experienced drug-induced hepatotoxicity. The development and validation of the iPSC-derived hepatocytes as an in vitro cell-based model of DILI is an essential first step in creating more predictive tools for understanding patient-specific hepatotoxic responses to drug treatment. In this study, we performed extensive morphological and functional analyses on iPSC-derived hepatocytes from a commercial source. iPSC-derived hepatocytes exhibit many of the key morphological and functional features of primary hepatocytes, including membrane polarity and production of glycogen, lipids, and key hepatic proteins, such as albumin, asialoglycoprotein receptor and α1-antitrypsin. They maintain functional activity for many drug-metabolizing enzyme pathways and possess active efflux capacity of marker substrates into bile canalicular compartments. Whole genome-wide array analysis of multiple batches of iPSC-derived cells showed that their transcriptional profiles are more similar to those from neonatal and adult hepatocytes than those from fetal liver. Results from experiments using prototype DILI compounds, such as acetaminophen and trovafloxacin, indicate that these cells are able to reproduce key characteristic metabolic and adaptive responses attributed to the drug-induced hepatotoxic effects in vivo. Overall, this novel system represents a promising new tool for understanding the underlying mechanisms of idiosyncratic DILI and for screening new compounds for DILI-related liabilities.
Collapse
Affiliation(s)
- Jingtao Lu
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709
| | | | - Lata Venkatarangan
- QPS Hepatic Biosciences, Research Triangle Park, North Carolina 27709; and
| | - Manda Miller
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709
| | - David A Mann
- QPS Hepatic Biosciences, Research Triangle Park, North Carolina 27709; and
| | - Paul B Watkins
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709; Schools of Medicine, Pharmacy and Public Health, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599
| | - Edward LeCluyse
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709;
| |
Collapse
|
28
|
Baxter M, Withey S, Harrison S, Segeritz CP, Zhang F, Atkinson-Dell R, Rowe C, Gerrard DT, Sison-Young R, Jenkins R, Henry J, Berry AA, Mohamet L, Best M, Fenwick SW, Malik H, Kitteringham NR, Goldring CE, Piper Hanley K, Vallier L, Hanley NA. Phenotypic and functional analyses show stem cell-derived hepatocyte-like cells better mimic fetal rather than adult hepatocytes. J Hepatol 2015; 62:581-9. [PMID: 25457200 PMCID: PMC4334496 DOI: 10.1016/j.jhep.2014.10.016] [Citation(s) in RCA: 240] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 09/18/2014] [Accepted: 10/09/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Hepatocyte-like cells (HLCs), differentiated from pluripotent stem cells by the use of soluble factors, can model human liver function and toxicity. However, at present HLC maturity and whether any deficit represents a true fetal state or aberrant differentiation is unclear and compounded by comparison to potentially deteriorated adult hepatocytes. Therefore, we generated HLCs from multiple lineages, using two different protocols, for direct comparison with fresh fetal and adult hepatocytes. METHODS Protocols were developed for robust differentiation. Multiple transcript, protein and functional analyses compared HLCs to fresh human fetal and adult hepatocytes. RESULTS HLCs were comparable to those of other laboratories by multiple parameters. Transcriptional changes during differentiation mimicked human embryogenesis and showed more similarity to pericentral than periportal hepatocytes. Unbiased proteomics demonstrated greater proximity to liver than 30 other human organs or tissues. However, by comparison to fresh material, HLC maturity was proven by transcript, protein and function to be fetal-like and short of the adult phenotype. The expression of 81% phase 1 enzymes in HLCs was significantly upregulated and half were statistically not different from fetal hepatocytes. HLCs secreted albumin and metabolized testosterone (CYP3A) and dextrorphan (CYP2D6) like fetal hepatocytes. In seven bespoke tests, devised by principal components analysis to distinguish fetal from adult hepatocytes, HLCs from two different source laboratories consistently demonstrated fetal characteristics. CONCLUSIONS HLCs from different sources are broadly comparable with unbiased proteomic evidence for faithful differentiation down the liver lineage. This current phenotype mimics human fetal rather than adult hepatocytes.
Collapse
Affiliation(s)
- Melissa Baxter
- Centre for Endocrinology & Diabetes, Institute of Human Development, Faculty of Medical & Human Sciences, University of Manchester, Manchester Academic Health Science Centre, AV Hill Building, Oxford Road, Manchester, UK
| | - Sarah Withey
- Centre for Endocrinology & Diabetes, Institute of Human Development, Faculty of Medical & Human Sciences, University of Manchester, Manchester Academic Health Science Centre, AV Hill Building, Oxford Road, Manchester, UK
| | - Sean Harrison
- Centre for Endocrinology & Diabetes, Institute of Human Development, Faculty of Medical & Human Sciences, University of Manchester, Manchester Academic Health Science Centre, AV Hill Building, Oxford Road, Manchester, UK
| | - Charis-Patricia Segeritz
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Institute for Regenerative Medicine, Department of Surgery, Robinson Way, Cambridge CB2 0SZ, UK; Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Fang Zhang
- Department of Pharmacology & Therapeutics and MRC Centre for Drug Safety Science, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, UK
| | - Rebecca Atkinson-Dell
- Centre for Endocrinology & Diabetes, Institute of Human Development, Faculty of Medical & Human Sciences, University of Manchester, Manchester Academic Health Science Centre, AV Hill Building, Oxford Road, Manchester, UK
| | - Cliff Rowe
- Centre for Endocrinology & Diabetes, Institute of Human Development, Faculty of Medical & Human Sciences, University of Manchester, Manchester Academic Health Science Centre, AV Hill Building, Oxford Road, Manchester, UK; Department of Pharmacology & Therapeutics and MRC Centre for Drug Safety Science, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, UK
| | - Dave T Gerrard
- Centre for Endocrinology & Diabetes, Institute of Human Development, Faculty of Medical & Human Sciences, University of Manchester, Manchester Academic Health Science Centre, AV Hill Building, Oxford Road, Manchester, UK; Bioinformatics, Faculty of Life Sciences, Michael Smith Building, Oxford Road, Manchester, UK
| | - Rowena Sison-Young
- Department of Pharmacology & Therapeutics and MRC Centre for Drug Safety Science, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, UK
| | - Roz Jenkins
- Department of Pharmacology & Therapeutics and MRC Centre for Drug Safety Science, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, UK
| | - Joanne Henry
- Department of Pharmacology & Therapeutics and MRC Centre for Drug Safety Science, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, UK
| | - Andrew A Berry
- Centre for Endocrinology & Diabetes, Institute of Human Development, Faculty of Medical & Human Sciences, University of Manchester, Manchester Academic Health Science Centre, AV Hill Building, Oxford Road, Manchester, UK
| | - Lisa Mohamet
- Stem Cell Research Group, Faculty of Medical & Human Sciences, University of Manchester, Manchester Academic Health Science Centre, AV Hill Building, Oxford Road, Manchester, UK
| | - Marie Best
- Human Genetics Division, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, UK
| | - Stephen W Fenwick
- North Western Hepatobiliary Unit, Aintree University Hospital NHS Foundation Trust, Longmoor Lane, Liverpool L9 7AL, UK
| | - Hassan Malik
- North Western Hepatobiliary Unit, Aintree University Hospital NHS Foundation Trust, Longmoor Lane, Liverpool L9 7AL, UK
| | - Neil R Kitteringham
- Department of Pharmacology & Therapeutics and MRC Centre for Drug Safety Science, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, UK
| | - Chris E Goldring
- Department of Pharmacology & Therapeutics and MRC Centre for Drug Safety Science, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, UK
| | - Karen Piper Hanley
- Centre for Endocrinology & Diabetes, Institute of Human Development, Faculty of Medical & Human Sciences, University of Manchester, Manchester Academic Health Science Centre, AV Hill Building, Oxford Road, Manchester, UK
| | - Ludovic Vallier
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Institute for Regenerative Medicine, Department of Surgery, Robinson Way, Cambridge CB2 0SZ, UK; Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Neil A Hanley
- Centre for Endocrinology & Diabetes, Institute of Human Development, Faculty of Medical & Human Sciences, University of Manchester, Manchester Academic Health Science Centre, AV Hill Building, Oxford Road, Manchester, UK; Endocrinology Department, Central Manchester University Hospitals NHS Foundation Trust, Grafton St, Manchester, UK.
| |
Collapse
|
29
|
van Wenum M, Chamuleau RAFM, van Gulik TM, Siliakus A, Seppen J, Hoekstra R. Bioartificial liversin vitroandin vivo: tailoring biocomponents to the expanding variety of applications. Expert Opin Biol Ther 2014; 14:1745-60. [DOI: 10.1517/14712598.2014.950651] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
30
|
Fuchs H, Theuser M, Wruck W, Adjaye J. miR-27 negatively regulates pluripotency-associated genes in human embryonal carcinoma cells. PLoS One 2014; 9:e111637. [PMID: 25369332 PMCID: PMC4219743 DOI: 10.1371/journal.pone.0111637] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 10/03/2014] [Indexed: 02/07/2023] Open
Abstract
Human embryonic stem cells and human embryonal carcinoma cells have been studied extensively with respect to the transcription factors (OCT4, SOX2 and NANOG), epigenetic modulators and associated signalling pathways that either promote self-renewal or induce differentiation in these cells. The ACTIVIN/NODAL axis (SMAD2/3) of the TGFß signalling pathway coupled with FGF signalling maintains self-renewal in these cells, whilst the BMP (SMAD1,5,8) axis promotes differentiation. Here we show that miR-27, a somatic-enriched miRNA, is activated upon RNAi-mediated suppression of OCT4 function in human embryonic stem cells. We further demonstrate that miR-27 negatively regulates the expression of the pluripotency-associated ACTIVIN/NODAL axis (SMAD2/3) of the TGFß signalling pathway by targeting ACVR2A, TGFßR1 and SMAD2. Additionally, we have identified a number of pluripotency-associated genes such as NANOG, LIN28, POLR3G and NR5A2 as novel miR-27 targets. Transcriptome analysis revealed that miR-27 over-expression in human embryonal carcinoma cells leads indeed to a significant up-regulation of genes involved in developmental pathways such as TGFß- and WNT-signalling.
Collapse
Affiliation(s)
- Heiko Fuchs
- Institute for Stem Cell Research and Regenerative Medicine, Faculty of Medicine, Heinrich Heine University, Duesseldorf, Germany
| | - Matthias Theuser
- Department of Vertebrate Genomics, Molecular Embryology and Aging Group, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Wasco Wruck
- Institute for Stem Cell Research and Regenerative Medicine, Faculty of Medicine, Heinrich Heine University, Duesseldorf, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Faculty of Medicine, Heinrich Heine University, Duesseldorf, Germany
- Department of Vertebrate Genomics, Molecular Embryology and Aging Group, Max Planck Institute for Molecular Genetics, Berlin, Germany
- * E-mail:
| |
Collapse
|
31
|
Krueger W, Boelsterli UA, Rasmussen TP. Stem Cell Strategies to Evaluate Idiosyncratic Drug-induced Liver Injury. J Clin Transl Hepatol 2014; 2:143-52. [PMID: 26355943 PMCID: PMC4521249 DOI: 10.14218/jcth.2014.00012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/13/2014] [Accepted: 06/07/2014] [Indexed: 12/14/2022] Open
Abstract
The host-dependent nature of idiosyncratic drug-induced liver injury (iDILI) suggests that rare genetic polymorphisms may contribute to the disease. Indeed, a few mutations in key genes have already been identified using conventional human genetics approaches. Over 50 commonly used drugs can precipitate iDILI, making this a substantial medical problem. Only recently have human induced pluripotent stem cells been used as a research tool to discover novel iDILI genes and to study the mechanisms of iDILI in vitro. Here we review the current state of stem cell use in the investigation of iDILI, with a special focus on genetics. In addition, the concerns and difficulties associated with genetics and animal model research are discussed. We then present the features of patient-specific pluripotent stem cells (which may be derived from iDILI patients themselves), and explain why these cells may be of great utility. A variety of recent approaches to produce hepatocyte-like cells from pluripotent cells and the associated advantages and limitations of such cells are discussed. Future directions for the use of stem cell science to investigate iDILI include novel ways to identify new iDILI genes, a consideration of epigenetic impacts on iDILI, and the development of new and improved strategies for the production of hepatocytes from human pluripotent cells.
Collapse
Affiliation(s)
- Winfried Krueger
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Urs A. Boelsterli
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Theodore P. Rasmussen
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
- University of Connecticut Stem Cell Institute, Storrs/Farmington, CT, USA
- Correspondence to: Theodore P. Rasmussen, Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, U-3092, Storrs, CT 06269, USA. Tel: +86-486-8339, Fax: +86-486-5792. E-mail:
| |
Collapse
|
32
|
Matz P, Adjaye J. Characterisation of human induced pluripotent stem cell-derived hepatocyte-like cells and endodermal progenitors. Eur J Med Res 2014. [PMCID: PMC4118433 DOI: 10.1186/2047-783x-19-s1-s8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
33
|
Fontana RJ. Pathogenesis of idiosyncratic drug-induced liver injury and clinical perspectives. Gastroenterology 2014; 146:914-28. [PMID: 24389305 PMCID: PMC4031195 DOI: 10.1053/j.gastro.2013.12.032] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 12/03/2013] [Accepted: 12/11/2013] [Indexed: 12/13/2022]
Abstract
Idiosyncratic drug-induced liver injury (DILI) is a rare disease that develops independently of drug dose, route, or duration of administration. Furthermore, idiosyncratic DILI is not a single disease entity but rather a spectrum of rare diseases with varying clinical, histological, and laboratory features. The pathogenesis of DILI is not fully understood. Standardization of the DILI nomenclature and methods to assess causality, along with the information provided by the LiverTox Web site, will harmonize and accelerate research on DILI. Studies of new serum biomarkers such as glutamate dehydrogenase, high mobility group box protein 1, and microRNA-122 could provide information for use in diagnosis and prognosis and provide important insights into the mechanisms of the pathogenesis of DILI. Single nucleotide polymorphisms in the HLA region have been associated with idiosyncratic hepatotoxicity attributed to flucloxacillin, ximelagatran, lapatinib, and amoxicillin-clavulanate. However, genome-wide association studies of pooled cases have not associated any genetic factors with idiosyncratic DILI. Whole genome and whole exome sequencing analyses are under way to study cases of DILI attributed to a single medication. Serum proteomic, transcriptome, and metabolome as well as intestinal microbiome analyses will increase our understanding of the mechanisms of this disorder. Further improvements to in vitro and in vivo test systems should advance our understanding of the causes, risk factors, and mechanisms of idiosyncratic DILI.
Collapse
|
34
|
Gerbal-Chaloin S, Funakoshi N, Caillaud A, Gondeau C, Champon B, Si-Tayeb K. Human induced pluripotent stem cells in hepatology: beyond the proof of concept. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 184:332-47. [PMID: 24269594 DOI: 10.1016/j.ajpath.2013.09.026] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/20/2013] [Accepted: 09/26/2013] [Indexed: 02/08/2023]
Abstract
The discovery of the wide plasticity of most cell types means that it is now possible to produce virtually any cell type in vitro. This concept, developed because of the possibility of reprogramming somatic cells toward induced pluripotent stem cells, provides the opportunity to produce specialized cells that harbor multiple phenotypical traits, thus integrating genetic interindividual variability. The field of hepatology has exploited this concept, and hepatocyte-like cells can now be differentiated from induced pluripotent stem cells. This review discusses the choice of somatic cells to be reprogrammed by emergent new and nonintegrative strategies, as well as the application of differentiated human induced pluripotent stem cells in hepatology, including liver development, disease modeling, host-pathogen interactions, and drug metabolism and toxicity. The actual consensus is that hepatocyte-like cells generated in vitro present an immature phenotype. Currently, developed strategies used to resolve this problem, such as overexpression of transcription factors, mimicking liver neonatal and postnatal modifications, and re-creating the three-dimensional hepatocyte environment in vitro and in vivo, are also discussed.
Collapse
Affiliation(s)
- Sabine Gerbal-Chaloin
- INSERM, U1087, Montpellier, France; UMR 1040, Université Montpellier 1, Montpellier, France
| | - Natalie Funakoshi
- INSERM, U1087, Montpellier, France; UMR 1040, Université Montpellier 1, Montpellier, France; Hepato-Gastroenterology Service B, Saint Eloi Hospital, CHU Montpellier, Montpellier, France
| | - Amandine Caillaud
- INSERM, UMR 1087, the Institute of the Thorax, Nantes, France; CNRS, UMR 6291, Nantes, France; School of Medicine, University of Nantes, Nantes, France
| | - Claire Gondeau
- INSERM, U1087, Montpellier, France; UMR 1040, Université Montpellier 1, Montpellier, France
| | - Benoite Champon
- INSERM, UMR 1087, the Institute of the Thorax, Nantes, France; CNRS, UMR 6291, Nantes, France; School of Medicine, University of Nantes, Nantes, France
| | - Karim Si-Tayeb
- INSERM, UMR 1087, the Institute of the Thorax, Nantes, France; CNRS, UMR 6291, Nantes, France; School of Medicine, University of Nantes, Nantes, France.
| |
Collapse
|
35
|
Kia R, Sison RLC, Heslop J, Kitteringham NR, Hanley N, Mills JS, Park BK, Goldring CEP. Stem cell-derived hepatocytes as a predictive model for drug-induced liver injury: are we there yet? Br J Clin Pharmacol 2013; 75:885-96. [PMID: 22703588 DOI: 10.1111/j.1365-2125.2012.04360.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 06/10/2012] [Indexed: 12/22/2022] Open
Abstract
Amongst the different types of adverse drug reactions, drug-induced liver injury is the most prominent cause of patient morbidity and mortality. However, the current available hepatic model systems developed for evaluating safety have limited utility and relevance as they do not fully recapitulate a fully functional hepatocyte, and do not sufficiently represent the genetic polymorphisms present in the population. The rapidly advancing research in stem cells raises the possibility of using human pluripotent stem cells in bridging this gap. The generation of human induced pluripotent stem cells via reprogramming of mature human somatic cells may also allow for disease modelling in vitro for the purposes of assessing drug safety and toxicology. This would also allow for better understanding of disease processes and thus facilitate in the potential identification of novel therapeutic targets. This review will focus on the current state of effort to derive hepatocytes from human pluripotent stem cells for potential use in hepatotoxicity evaluation and aims to provide an insight as to where the future of the field may lie.
Collapse
Affiliation(s)
- Richard Kia
- Department of Molecular and Clinical Pharmacology, University of Liverpool, MRC Centre for Drug Safety Science, Liverpool, UK
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Al-Nbaheen M, Vishnubalaji R, Ali D, Bouslimi A, Al-Jassir F, Megges M, Prigione A, Adjaye J, Kassem M, Aldahmash A. Human stromal (mesenchymal) stem cells from bone marrow, adipose tissue and skin exhibit differences in molecular phenotype and differentiation potential. Stem Cell Rev Rep 2013; 9:32-43. [PMID: 22529014 PMCID: PMC3563956 DOI: 10.1007/s12015-012-9365-8] [Citation(s) in RCA: 261] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human stromal (mesenchymal) stem cells (hMSCs) are multipotent stem cells with ability to differentiate into mesoderm-type cells e.g. osteoblasts and adipocytes and thus they are being introduced into clinical trials for tissue regeneration. Traditionally, hMSCs have been isolated from bone marrow, but the number of cells obtained is limited. Here, we compared the MSC-like cell populations, obtained from alternative sources for MSC: adipose tissue and skin, with the standard phenotype of human bone marrow MSC (BM-MSCs). MSC from human adipose tissue (human adipose stromal cells (hATSCs)) and human skin (human adult skin stromal cells, (hASSCs) and human new-born skin stromal cells (hNSSCs)) grew readily in culture and the growth rate was highest in hNSSCs and lowest in hATSCs. Compared with phenotype of hBM-MSC, all cell populations were CD34−, CD45−, CD14−, CD31−, HLA-DR−, CD13+, CD29+, CD44+, CD73+, CD90+,and CD105+. When exposed to in vitro differentiation, hATSCs, hASSCs and hNSSCs exhibited quantitative differences in their ability to differentiate into adipocytes and to osteoblastic cells. Using a microarray-based approach we have unveiled a common MSC molecular signature composed of 33 CD markers including known MSC markers and several novel markers e.g. CD165, CD276, and CD82. However, significant differences in the molecular phenotype between these different stromal cell populations were observed suggesting ontological and functional differences. In conclusion, MSC populations obtained from different tissues exhibit significant differences in their proliferation, differentiation and molecular phenotype, which should be taken into consideration when planning their use in clinical protocols.
Collapse
Affiliation(s)
- May Al-Nbaheen
- Stem Cell Unit, Department of Anatomy 28, College of Medicine, King Saud University, P.O. Box 2925, Riyadh, 11461, Kingdom of Saudi Arabia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Rao MS, Sasikala M, Reddy DN. Thinking outside the liver: induced pluripotent stem cells for hepatic applications. World J Gastroenterol 2013; 19:3385-96. [PMID: 23801830 PMCID: PMC3683676 DOI: 10.3748/wjg.v19.i22.3385] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 12/06/2011] [Accepted: 12/15/2011] [Indexed: 02/06/2023] Open
Abstract
The discovery of induced pluripotent stem cells (iPSCs) unraveled a mystery in stem cell research, after identification of four re-programming factors for generating pluripotent stem cells without the need of embryos. This breakthrough in generating iPSCs from somatic cells has overcome the ethical issues and immune rejection involved in the use of human embryonic stem cells. Hence, iPSCs form a great potential source for developing disease models, drug toxicity screening and cell-based therapies. These cells have the potential to differentiate into desired cell types, including hepatocytes, under in vitro as well as under in vivo conditions given the proper microenvironment. iPSC-derived hepatocytes could be useful as an unlimited source, which can be utilized in disease modeling, drug toxicity testing and producing autologous cell therapies that would avoid immune rejection and enable correction of gene defects prior to cell transplantation. In this review, we discuss the induction methods, role of reprogramming factors, and characterization of iPSCs, along with hepatocyte differentiation from iPSCs and potential applications. Further, we discuss the location and detection of liver stem cells and their role in liver regeneration. Although tumor formation and genetic mutations are a cause of concern, iPSCs still form a promising source for clinical applications.
Collapse
|
38
|
Ji S, Zhang L, Hui L. Cell fate conversion: direct induction of hepatocyte-like cells from fibroblasts. J Cell Biochem 2013; 114:256-65. [PMID: 22948752 DOI: 10.1002/jcb.24380] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 08/27/2012] [Indexed: 12/17/2022]
Abstract
One of the essential features of stem cells is their cellular plasticity to differentiate into daughter cells with defined functions. Recently, induction of pluripotent stem cells from somatic cells by defined transcription factors led to the focus on cellular plasticity of terminally differentiated cells. This approach is adopted by other studies to demonstrate the cell fate conversion between different lineages of terminally differentiated cells. We and others showed that induced hepatocyte-like (iHep) cells are directly converted from mouse fibroblasts by overexpression of liver-enriched transcription factors. iHep cells as well as pluripotent stem cell- or mesenchymal stem cell-derived hepatocyte-like cells provide potential cell sources for disease modeling, transplantation, and tissue engineering independent of donor organs. Here, we review the latest advances in generating hepatocyte-like cells and summarize general criteria for evaluating these cells. In addition, we propose a possible role of the p19(Arf) /p53 pathway in cell fate maintenance, which apparently limits the formation of induced pluripotent stem (iPS) cells and iHep cells.
Collapse
Affiliation(s)
- Shuyi Ji
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Yueyang Road 320, Shanghai 200031, China
| | | | | |
Collapse
|
39
|
Identification of small molecules for human hepatocyte expansion and iPS differentiation. Nat Chem Biol 2013; 9:514-20. [PMID: 23728495 PMCID: PMC3720805 DOI: 10.1038/nchembio.1270] [Citation(s) in RCA: 208] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 05/01/2013] [Indexed: 12/21/2022]
Abstract
Cell-based therapies hold the potential to alleviate the growing burden of liver diseases. Such therapies require human hepatocytes, which, within the stromal context of the liver, are capable of many rounds of replication. However, this ability is lost ex vivo, and human hepatocyte sourcing has limited many fields of research for decades. Here we developed a high-throughput screening platform for primary human hepatocytes to identify small molecules in two different classes that can be used to generate renewable sources of functional human hepatocytes. The first class induced functional proliferation of primary human hepatocytes in vitro. The second class enhanced hepatocyte functions and promoted the differentiation of induced pluripotent stem cell-derived hepatocytes toward a more mature phenotype than what was previously obtainable. The identification of these small molecules can help address a major challenge affecting many facets of liver research and may lead to the development of new therapeutics for liver diseases.
Collapse
|
40
|
Human induced pluripotent stem cells and their use in drug discovery for toxicity testing. Toxicol Lett 2013; 219:49-58. [PMID: 23470867 DOI: 10.1016/j.toxlet.2013.02.020] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 02/21/2013] [Accepted: 02/23/2013] [Indexed: 12/23/2022]
Abstract
Predicting human safety risks of novel xenobiotics remains a major challenge, partly due to the limited availability of human cells to evaluate tissue-specific toxicity. Recent progress in the production of human induced pluripotent stem cells (hiPSCs) may fill this gap. hiPSCs can be continuously expanded in culture in an undifferentiated state and then differentiated to form most cell types. Thus, it is becoming technically feasible to generate large quantities of human cell types and, in combination with relatively new detection methods, to develop higher-throughput in vitro assays that quantify tissue-specific biological properties. Indeed, the first wave of large scale hiSC-differentiated cell types including patient-derived hiPSCS are now commercially available. However, significant improvements in hiPSC production and differentiation processes are required before cell-based toxicity assays that accurately reflect mature tissue phenotypes can be delivered and implemented in a cost-effective manner. In this review, we discuss the promising alignment of hiPSCs and recently emerging technologies to quantify tissue-specific functions. We emphasize liver, cardiovascular, and CNS safety risks and highlight limitations that must be overcome before routine screening for toxicity pathways in hiSC-derived cells can be established.
Collapse
|
41
|
Pomp O, Colman A. Disease modelling using induced pluripotent stem cells: status and prospects. Bioessays 2012; 35:271-80. [PMID: 23148027 DOI: 10.1002/bies.201200088] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The ability to convert human somatic cells into induced pluripotent stem cells (iPSCs) is allowing the production of custom-tailored cells for drug discovery and for the study of disease phenotypes at the cellular and molecular level. IPSCs have been derived from patients suffering from a large variety of disorders with different severities. In many cases, disease related phenotypes have been observed in iPSCs or their lineage-specific progeny. Several proof of concept studies have demonstrated that these phenotypes can be reversed in vitro using approved drugs. However, several challenges must be overcome to take full advantage of this technology. Here, we highlight recent advances in the field and discuss the main challenges associated with this technology as it applies to disease modelling.
Collapse
Affiliation(s)
- Oz Pomp
- Institute of Medical Biology, #06-06 Immunos, Singapore
| | | |
Collapse
|
42
|
Hua M, Zhang W, Li W, Li X, Liu B, Lu X, Zhang H. Molecular mechanisms regulating the establishment of hepatocyte polarity during human hepatic progenitor cell differentiation into a functional hepatocyte-like phenotype. J Cell Sci 2012; 125:5800-10. [PMID: 22976305 DOI: 10.1242/jcs.110551] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The correct functioning of hepatocytes requires the establishment and maintenance of hepatocyte polarity. However, the mechanisms regulating the generation of hepatocyte polarity are not completely understood. The differentiation of human fetal hepatic progenitor cells (hFHPCs) into functional hepatocytes provides a powerful in vitro model system for studying the molecular mechanisms governing hepatocyte development. In this study, we used a two-stage differentiation protocol to generate functional polarized hepatocyte-like cells (HLCs) from hFHPCs. Global gene expression profiling was performed on triplicate samples of hFHPCs, immature-HLCs and mature-HLCs. When the differential gene expression was compared based on the differentiation stage, a number of genes were identified that might be essential for establishing and maintaining hepatocyte polarity. These genes include those that encode actin filament-binding protein, protein tyrosine kinase activity molecules, and components of signaling pathways, such as PTK7, PARD3, PRKCI and CDC42. Based on known and predicted protein-protein interactions, the candidate genes were assigned to networks and clustered into functional categories. The expression of several of these genes was confirmed using real-time RT-PCR. By inactivating genes using small interfering RNA, we demonstrated that PTK7 and PARD3 promote hepatic polarity formation and affect F-actin organization. These results provide unique insight into the complex process of polarization during hepatocyte differentiation, indicating key genes and signaling molecules governing hepatocyte differentiation.
Collapse
Affiliation(s)
- Mingxi Hua
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, 100069, China
| | | | | | | | | | | | | |
Collapse
|
43
|
Jozefczuk J, Kashofer K, Ummanni R, Henjes F, Rehman S, Geenen S, Wruck W, Regenbrecht C, Daskalaki A, Wierling C, Turano P, Bertini I, Korf U, Zatloukal K, Westerhoff HV, Lehrach H, Adjaye J. A Systems Biology Approach to Deciphering the Etiology of Steatosis Employing Patient-Derived Dermal Fibroblasts and iPS Cells. Front Physiol 2012; 3:339. [PMID: 22969728 PMCID: PMC3432516 DOI: 10.3389/fphys.2012.00339] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 08/03/2012] [Indexed: 12/23/2022] Open
Abstract
Non-alcoholic fatty liver disease comprises a broad spectrum of disease states ranging from simple steatosis to non-alcoholic steatohepatitis. As a result of increases in the prevalences of obesity, insulin resistance, and hyperlipidemia, the number of people with hepatic steatosis continues to increase. Differences in susceptibility to steatohepatitis and its progression to cirrhosis have been attributed to a complex interplay of genetic and external factors all addressing the intracellular network. Increase in sugar or refined carbohydrate consumption results in an increase of insulin and insulin resistance that can lead to the accumulation of fat in the liver. Here we demonstrate how a multidisciplinary approach encompassing cellular reprogramming, transcriptomics, proteomics, metabolomics, modeling, network reconstruction, and data management can be employed to unveil the mechanisms underlying the progression of steatosis. Proteomics revealed reduced AKT/mTOR signaling in fibroblasts derived from steatosis patients and further establishes that the insulin-resistant phenotype is present not only in insulin-metabolizing central organs, e.g., the liver, but is also manifested in skin fibroblasts. Transcriptome data enabled the generation of a regulatory network based on the transcription factor SREBF1, linked to a metabolic network of glycerolipid, and fatty acid biosynthesis including the downstream transcriptional targets of SREBF1 which include LIPIN1 (LPIN) and low density lipoprotein receptor. Glutathione metabolism was among the pathways enriched in steatosis patients in comparison to healthy controls. By using a model of the glutathione pathway we predict a significant increase in the flux through glutathione synthesis as both gamma-glutamylcysteine synthetase and glutathione synthetase have an increased flux. We anticipate that a larger cohort of patients and matched controls will confirm our preliminary findings presented here.
Collapse
Affiliation(s)
- Justyna Jozefczuk
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Drews K, Jozefczuk J, Prigione A, Adjaye J. Human induced pluripotent stem cells—from mechanisms to clinical applications. J Mol Med (Berl) 2012; 90:735-45. [PMID: 22643868 DOI: 10.1007/s00109-012-0913-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 05/03/2012] [Accepted: 05/06/2012] [Indexed: 01/30/2023]
Affiliation(s)
- Katharina Drews
- Molecular Embryology and Aging Group, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Ihnestr. 63-73 14195, Berlin, Germany
| | | | | | | |
Collapse
|
45
|
Zhou WL, Medine CN, Zhu L, Hay DC. Stem cell differentiation and human liver disease. World J Gastroenterol 2012; 18:2018-25. [PMID: 22563188 PMCID: PMC3342599 DOI: 10.3748/wjg.v18.i17.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 02/08/2012] [Accepted: 02/26/2012] [Indexed: 02/06/2023] Open
Abstract
Human stem cells are scalable cell populations capable of cellular differentiation. This makes them a very attractive in vitro cellular resource and in theory provides unlimited amounts of primary cells. Such an approach has the potential to improve our understanding of human biology and treating disease. In the future it may be possible to deploy novel stem cell-based approaches to treat human liver diseases. In recent years, efficient hepatic differentiation from human stem cells has been achieved by several research groups including our own. In this review we provide an overview of the field and discuss the future potential and limitations of stem cell technology.
Collapse
|
46
|
Ho PJ, Yen ML, Yet SF, Yen BL. Current Applications of Human Pluripotent Stem Cells: Possibilities and Challenges. Cell Transplant 2012; 21:801-14. [DOI: 10.3727/096368911x627507] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Stem cells are self-renewable cells with the differentiation capacity to develop into somatic cells with biological functions. This ability to sustain a renewable source of multi- and/or pluripotential differentiation has brought new hope to the field of regenerative medicine in terms of cell therapy and tissue engineering. Moreover, stem cells are invaluable tools as in vitro models for studying diverse fields, from basic scientific questions such as developmental processes and lineage commitment, to practical application including drug screening and testing. The stem cells with widest differentiation potential are pluripotent stem cells (PSCs), which are rare cells with the ability to generate somatic cells from all three germ layers. PSCs are considered the most optimal choice for therapeutic potential of stem cells, bringing new impetus to the field of regenerative medicine. In this article, we discuss the therapeutic potential of human PSCs (hPSCs) including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), reviewing the current preclinical and clinical data using these stem cells. We describe the classification of different sources of hPSCs, ongoing research, and currently encountered clinical obstacles of these novel and versatile human stem cells.
Collapse
Affiliation(s)
- Pai-Jiun Ho
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Men-Luh Yen
- Departmant of Primary Medicine and Department of Obstetrics/Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shaw-Fang Yet
- Cardiovascular Research Group, Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - B. Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| |
Collapse
|
47
|
Forbes SJ, Newsome PN. New horizons for stem cell therapy in liver disease. J Hepatol 2012; 56:496-9. [PMID: 21798218 DOI: 10.1016/j.jhep.2011.06.022] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 06/20/2011] [Accepted: 06/20/2011] [Indexed: 12/15/2022]
Abstract
There is an increasing range of potential applications of stem cells in liver diseases, with many clinical studies already undertaken. We identify four of the main areas which we propose stem cell therapy could be a realistic aim for in the future: (1) to improve regeneration and reduce scarring in liver cirrhosis by modulating the liver's own regenerative processes, (2) to down-regulate immune mediated liver damage, (3) supplying hepatocyte-like cells (HLCs) derived from stem cells for use in extracorporeal bio-artificial liver machines, and (4) to use stem cell derived HLCs for cell transplantation to supplement or replace hepatocyte function.
Collapse
Affiliation(s)
- Stuart J Forbes
- MRC Centre for Regenerative Medicine and MRC, Edinburgh University, UK.
| | | |
Collapse
|
48
|
Kim TI. [Clinical trials with stem cells in digestive diseases and future perspectives]. THE KOREAN JOURNAL OF GASTROENTEROLOGY 2012; 58:139-43. [PMID: 21960101 DOI: 10.4166/kjg.2011.58.3.139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Many techniques for isolation, expansion and handling of stem cells are being developed rapidly, and preclinical evidence has shown the possibility to use this technology for refractory diseases in the near future. Among refractory digestive diseases, Crohn's disease and liver cirrhosis may be two main diseases where stem cell therapy can be applied for anti-inflammation and regeneration of tissue. Currently, with respect to these two diseases, clinical trials using hematopoietic stem cells and mesenchymal stem cells from bone marrow or adipose tissue have shown some evidence of clinical benefits to immune modulation, suppression of inflammation and regeneration of functional cells. However, for the development of practical stem cell therapy, we need more data on underlying mechanisms, effective subpopulation of stem cells and its sources, and effective parameters for monitoring and estimation. With technical advances, the research on embryonic and induced pluripotent stem cells will also contribute to the new therapeutic strategies for digestive regenerative medicine. In the future, a variety of stem cell therapies may be therapeutic options for refractory digestive diseases, but many technical challenges remain to be solved. (Korean J Gastroenterol 2011;58:139-143).
Collapse
Affiliation(s)
- Tae Il Kim
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
49
|
Pistollato F, Bremer-Hoffmann S, Healy L, Young L, Stacey G. Standardization of pluripotent stem cell cultures for toxicity testing. Expert Opin Drug Metab Toxicol 2012; 8:239-57. [PMID: 22248265 DOI: 10.1517/17425255.2012.639763] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Pluripotent stem cell (PSC) lines offer a unique opportunity to derive various human cell types that can be exploited for human safety assessments in vitro and as such contribute to modern mechanistically oriented toxicity testing. AREAS COVERED This article reviews the two major types of PSC cultures that are currently most promising for toxicological applications: human embryonic stem cell lines and human induced PSC lines. Through the review, the article explains how these cell types will improve the current safety evaluations of chemicals and will allow a more efficient selection of drug candidates. Additionally, the article discusses the important issues of maintaining PSCs as well as their differentiation efficiency. EXPERT OPINION The demonstration of the reliability and relevance of in vitro toxicity tests for a given purpose is mandatory for their use in regulatory toxicity testing. Given the peculiar nature of PSCs, a high level of standardization of undifferentiated cell cultures as well as of the differentiation process is required in order to ensure the establishment of robust test systems. It is, therefore, of pivotal importance to define and internationally agree on crucial parameters to judge the quality of the cellular models before enrolling them for toxicity testing.
Collapse
Affiliation(s)
- Francesca Pistollato
- Institute for Health & Consumer Protection, Systems Toxicology Unit, Joint Research Centre, European Commission, Ispra, Italy
| | | | | | | | | |
Collapse
|
50
|
Sison-Young RLC, Kia R, Heslop J, Kelly L, Rowe C, Cross MJ, Kitteringham NR, Hanley N, Park BK, Goldring CEP. Human pluripotent stem cells for modeling toxicity. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2012; 63:207-256. [PMID: 22776643 DOI: 10.1016/b978-0-12-398339-8.00006-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The development of xenobiotics, driven by the demand for therapeutic, domestic and industrial uses continues to grow. However, along with this increasing demand is the risk of xenobiotic-induced toxicity. Currently, safety screening of xenobiotics uses a plethora of animal and in vitro model systems which have over the decades proven useful during compound development and for application in mechanistic studies of xenobiotic-induced toxicity. However, these assessments have proven to be animal-intensive and costly. More importantly, the prevalence of xenobiotic-induced toxicity is still significantly high, causing patient morbidity and mortality, and a costly impediment during drug development. This suggests that the current models for drug safety screening are not reliable in toxicity prediction, and the results not easily translatable to the clinic due to insensitive assays that do not recapitulate fully the complex phenotype of a functional cell type in vivo. Recent advances in the field of stem cell research have potentially allowed for a readily available source of metabolically competent cells for toxicity studies, derived using human pluripotent stem cells harnessed from embryos or reprogrammed from mature somatic cells. Pluripotent stem cell-derived cell types also allow for potential disease modeling in vitro for the purposes of drug toxicology and safety pharmacology, making this model possibly more predictive of drug toxicity compared with existing models. This article will review the advances and challenges of using human pluripotent stem cells for modeling metabolism and toxicity, and offer some perspectives as to where its future may lie.
Collapse
Affiliation(s)
- R L C Sison-Young
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|