1
|
Traini L, Negueruela J, Elvira B, St-Pierre-Wijckmans W, Vandenbempt V, Buss CE, Li A, Pérez-Chávez I, Ribeiro-Costa F, Nunes M, Messens J, Ezeriņa D, Hay DC, Bansal M, Gurzov EN. Genome editing of TXNIP in human pluripotent stem cells for the generation of hepatocyte-like cells and insulin-producing islet-like aggregates. Stem Cell Res Ther 2025; 16:225. [PMID: 40320524 PMCID: PMC12051322 DOI: 10.1186/s13287-025-04314-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Thioredoxin-interacting protein (TXNIP) plays a role in regulating endoplasmic reticulum (ER) and oxidative stress, which disrupt glucose homeostasis in diabetes. However, the impact of TXNIP deficiency on the differentiation and functionality of human stem cell-derived somatic metabolic cells remains unclear. METHODS We used CRISPR-Cas12a genome editing to generate TXNIP-deficient (TXNIP-/-) H1 human embryonic stem cells (H1-hESCs). These cells were differentiated into hepatocyte-like cells (HLCs) and stem-cell-derived insulin-producing islets (SC-islets). The maturation and functionality TXNIP-/- and TXNIP+/+ SC-islets were assessed by implantation under the kidney capsule of male or female NOD-SCID mice. RESULTS TXNIP deficiency significantly increased H1-hESC proliferation without affecting pluripotency, viability, or differentiation potential into HLCs and SC-islets. Bulk RNA-sequencing of thapsigargin-treated TXNIP-/- and TXNIP+/+ hESCs revealed differential expression of stress-responsive genes, with enriched apoptosis-related pathways in TXNIP+/+ cells, but minimal transcriptional changes specific to TXNIP deficiency. In HLCs, TXNIP deletion reduced albumin secretion and insulin signalling, as indicated by decreased AKT phosphorylation, while showing no differences in glycolytic activity or lipid metabolism markers. Under thapsigargin-induced ER stress, TXNIP-/- HLCs exhibited transiently reduced eIF2α phosphorylation and lower BiP expression, suggesting compromised adaptive responses to prolonged stress. SC-islets derived from TXNIP-/- hESCs showed comparable viability, endocrine cell composition, and cytokine responses to TXNIP+/+ islets. Following IFNα or IFNγ treatment, STAT1 phosphorylation was increased in TXNIP-/- SC-islets, indicating that IFN signalling remained intact despite TXNIP deficiency. Upon implantation into NOD-SCID mice, both TXNIP-/- and TXNIP+/+ SC-islets produced human C-peptide and responded to glucose stimulation. However, TXNIP-/- SC-islets did not demonstrate enhanced glycaemic control or glucose-stimulated insulin secretion compared to controls. CONCLUSIONS Our study demonstrates that TXNIP deficiency does not improve the differentiation or functionality of HLCs and SC-islets. We present the generation and characterisation of TXNIP-/- and TXNIP+/+ H1-hESCs, HLCs, and SC-islets as valuable models for future studies on the role of TXNIP in metabolic cell biology.
Collapse
Affiliation(s)
- Leonardo Traini
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Javier Negueruela
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Bernat Elvira
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Wadsen St-Pierre-Wijckmans
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Valerie Vandenbempt
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Carlos E Buss
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Ao Li
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Israel Pérez-Chávez
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
- VIB-VUB Center for Structural Biology, Vlaams Instituut Voor Biotechnologie, B-1050, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Francisco Ribeiro-Costa
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Mariana Nunes
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium
| | - Joris Messens
- VIB-VUB Center for Structural Biology, Vlaams Instituut Voor Biotechnologie, B-1050, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Daria Ezeriņa
- VIB-VUB Center for Structural Biology, Vlaams Instituut Voor Biotechnologie, B-1050, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - David C Hay
- Institute for Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Mayank Bansal
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium.
| | - Esteban N Gurzov
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Route de Lennik 808, B-1070, Brussels, Belgium.
| |
Collapse
|
2
|
Gilglioni EH, Li A, St-Pierre-Wijckmans W, Shen TK, Pérez-Chávez I, Hovhannisyan G, Lisjak M, Negueruela J, Vandenbempt V, Bauzá-Martinez J, Herranz JM, Ezeriņa D, Demine S, Feng Z, Vignane T, Otero Sanchez L, Lambertucci F, Prašnická A, Devière J, Hay DC, Encinar JA, Singh SP, Messens J, Filipovic MR, Sharpe HJ, Trépo E, Wu W, Gurzov EN. PTPRK regulates glycolysis and de novo lipogenesis to promote hepatocyte metabolic reprogramming in obesity. Nat Commun 2024; 15:9522. [PMID: 39496584 PMCID: PMC11535053 DOI: 10.1038/s41467-024-53733-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 10/22/2024] [Indexed: 11/06/2024] Open
Abstract
Fat accumulation, de novo lipogenesis, and glycolysis are key drivers of hepatocyte reprogramming and the consequent metabolic dysfunction-associated steatotic liver disease (MASLD). Here we report that obesity leads to dysregulated expression of hepatic protein-tyrosine phosphatases (PTPs). PTPRK was found to be increased in steatotic hepatocytes in both humans and mice, and correlates positively with PPARγ-induced lipogenic signaling. High-fat-fed PTPRK knockout male and female mice have lower weight gain and reduced hepatic fat accumulation. Phosphoproteomic analysis in primary hepatocytes and hepatic metabolomics identified fructose-1,6-bisphosphatase 1 and glycolysis as PTPRK targets in metabolic reprogramming. Mechanistically, PTPRK-induced glycolysis enhances PPARγ and lipogenesis in hepatocytes. Silencing PTPRK in liver cancer cell lines reduces colony-forming capacity and high-fat-fed PTPRK knockout mice exposed to a hepatic carcinogen develop smaller tumours. Our study defines the role of PTPRK in the regulation of hepatic glycolysis, lipid metabolism, and tumour development in obesity.
Collapse
Affiliation(s)
- Eduardo H Gilglioni
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, B-1070, Brussels, Belgium
| | - Ao Li
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, B-1070, Brussels, Belgium
| | | | - Tzu-Keng Shen
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, B-1070, Brussels, Belgium
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Israel Pérez-Chávez
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, B-1070, Brussels, Belgium
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Garnik Hovhannisyan
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, B-1070, Brussels, Belgium
| | - Michela Lisjak
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, B-1070, Brussels, Belgium
| | - Javier Negueruela
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, B-1070, Brussels, Belgium
| | - Valerie Vandenbempt
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, B-1070, Brussels, Belgium
| | - Julia Bauzá-Martinez
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH, Utrecht, The Netherlands
- Netherlands Proteomics Centre, 3584 CH, Utrecht, The Netherlands
| | - Jose M Herranz
- Hepatology Program, CIMA, University of Navarra, 31009, Pamplona, Spain
| | - Daria Ezeriņa
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Stéphane Demine
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, B-1070, Brussels, Belgium
| | - Zheng Feng
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, B-1070, Brussels, Belgium
| | - Thibaut Vignane
- Leibniz Institute for Analytical Sciences, ISAS e.V., 44139, Dortmund, Germany
| | - Lukas Otero Sanchez
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles, B-1070, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université libre de Bruxelles, B-1070, Brussels, Belgium
| | - Flavia Lambertucci
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, B-1070, Brussels, Belgium
| | - Alena Prašnická
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, B-1070, Brussels, Belgium
| | - Jacques Devière
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles, B-1070, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université libre de Bruxelles, B-1070, Brussels, Belgium
| | - David C Hay
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Jose A Encinar
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDIBE), 03202, Elche, Spain
| | - Sumeet Pal Singh
- IRIBHM, Université libre de Bruxelles, B-1070, Brussels, Belgium
| | - Joris Messens
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Milos R Filipovic
- Leibniz Institute for Analytical Sciences, ISAS e.V., 44139, Dortmund, Germany
| | - Hayley J Sharpe
- Signalling Programme, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Eric Trépo
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles, B-1070, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université libre de Bruxelles, B-1070, Brussels, Belgium
| | - Wei Wu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH, Utrecht, The Netherlands
- Netherlands Proteomics Centre, 3584 CH, Utrecht, The Netherlands
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
- Department of Pharmacy & Pharmaceutical Sciences, National University of Singapore, Singapore, 117543, Singapore
| | - Esteban N Gurzov
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, B-1070, Brussels, Belgium.
- WELBIO Department, WEL Research Institute, B-1300, Wavre, Belgium.
| |
Collapse
|
3
|
Shi Y, Deng J, Sang X, Wang Y, He F, Chen X, Xu A, Wu F. Generation of Hepatocytes and Nonparenchymal Cell Codifferentiation System from Human-Induced Pluripotent Stem Cells. Stem Cells Int 2022; 2022:3222427. [PMID: 36467281 PMCID: PMC9709383 DOI: 10.1155/2022/3222427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 09/22/2022] [Accepted: 10/13/2022] [Indexed: 02/05/2024] Open
Abstract
To date, hepatocytes derived from human-induced pluripotent stem cells (hiPSC) provide a potentially unlimited resource for clinical application and drug development. However, most hiPSC-derived hepatocyte-like cells initiated differentiation from highly purified definitive endoderm, which are insufficient to accurately replicate the complex regulation of signals among multiple cells and tissues during liver organogenesis, thereby displaying an immature phenotypic and short survival time in vitro. Here, we described a protocol to achieve codifferentiation of endoderm-derived hepatocytes and mesoderm-derived nonparenchymal cells by the inclusion of BMP4 into hepatic differentiation medium, which has a beneficial effect on the hepatocyte maturation and lifespan in vitro. Our codifferentiation system suggests the important role of nonparenchymal cells in liver organogenesis. Hopefully, these hepatocytes described here provide a promising approach in the therapy of liver diseases.
Collapse
Affiliation(s)
- Ying Shi
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, College of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jiali Deng
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, College of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiaopu Sang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yihang Wang
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, College of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Fei He
- Department of Central Laboratory, Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, China
| | - Xiaoni Chen
- Department of Central Laboratory, Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, China
| | - Anlong Xu
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, College of Life Sciences, Sun Yat-sen University, Guangzhou, China
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Fenfang Wu
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, College of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Department of Central Laboratory, Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
4
|
Li R, Zhao Y, Yourick JJ, Sprando RL, Gao X. Phenotypical, functional and transcriptomic comparison of two modified methods of hepatocyte differentiation from human induced pluripotent stem cells. Biomed Rep 2022; 16:43. [PMID: 35371477 PMCID: PMC8972237 DOI: 10.3892/br.2022.1526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Directed differentiation of human induced pluripotent stem cells (iPSCs) into hepatocytes could provide an unlimited source of liver cells, and therefore holds great promise for regenerative medicine, disease modeling, drug screening and toxicology studies. Various methods have been established during the past decade to differentiate human iPSCs into hepatocyte-like cells (HLCs) using growth factors and/or small molecules. However, direct comparison of the differentiation efficiency and the quality of the final HLCs between different methods has rarely been reported. In the current study, two hepatocyte differentiation methods were devised, termed Method 1 and 2, through modifying existing well-known hepatocyte differentiation strategies, and the resultant cells were compared phenotypically and functionally at different stages of hepatocyte differentiation. Compared to Method 1, higher differentiation efficiency and reproducibility were observed in Method 2, which generated highly homogeneous functional HLCs at the end of the differentiation process. The cells exhibited morphology closely resembling primary human hepatocytes and expressed high levels of hepatic protein markers. More importantly, these HLCs demonstrated several essential characteristics of mature hepatocytes, including major serum protein (albumin, fibronectin and α-1 antitrypsin) secretion, urea release, glycogen storage and inducible cytochrome P450 activity. Further transcriptomic comparison of the HLCs derived from the two methods identified 1,481 differentially expressed genes (DEGs); 290 Gene Ontology terms in the biological process category were enriched by these genes, which were further categorized into 34 functional classes. Pathway analysis of the DEGs identified several signaling pathways closely involved in hepatocyte differentiation of pluripotent stem cells, including 'signaling pathways regulating pluripotency of stem cells', 'Wnt signaling pathway', 'TGF-beta signaling pathway' and 'PI3K-Akt signaling pathway'. These results may provide a molecular basis for the differences observed between the two differentiation methods and suggest ways to further improve hepatocyte differentiation in order to obtain more mature HLCs for biomedical applications.
Collapse
Affiliation(s)
- Rong Li
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD 20708, USA
| | - Yang Zhao
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD 20708, USA
| | - Jeffrey J Yourick
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD 20708, USA
| | - Robert L Sprando
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD 20708, USA
| | - Xiugong Gao
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD 20708, USA
| |
Collapse
|
5
|
Telles-Silva KA, Pacheco L, Komatsu S, Chianca F, Caires-Júnior LC, Araujo BHS, Goulart E, Zatz M. Applied Hepatic Bioengineering: Modeling the Human Liver Using Organoid and Liver-on-a-Chip Technologies. Front Bioeng Biotechnol 2022; 10:845360. [PMID: 35237587 PMCID: PMC8882846 DOI: 10.3389/fbioe.2022.845360] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/19/2022] [Indexed: 12/19/2022] Open
Abstract
The liver is the most important metabolic hub of endo and xenobiotic compounds. Pre-clinical studies using rodents to evaluate the toxicity of new drugs and cosmetics may produce inconclusive results for predicting clinical outcomes in humans, moreover being banned in the European Union. Human liver modeling using primary hepatocytes presents low reproducibility due to batch-to-batch variability, while iPSC-derived hepatocytes in monolayer cultures (2D) show reduced cellular functionality. Here we review the current status of the two most robust in vitro approaches in improving hepatocyte phenotype and metabolism while mimicking the hepatic physiological microenvironment: organoids and liver-on-chip. Both technologies are reviewed in design and manufacturing techniques, following cellular composition and functionality. Furthermore, drug screening and liver diseases modeling efficiencies are summarized. Finally, organoid and liver-on-chip technologies are compared regarding advantages and limitations, aiming to guide the selection of appropriate models for translational research and the development of such technologies.
Collapse
Affiliation(s)
- Kayque Alves Telles-Silva
- Human Genome and Stem-Cell Research Center (HUG-CEL), Institute of Biosciences, University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Lara Pacheco
- Human Genome and Stem-Cell Research Center (HUG-CEL), Institute of Biosciences, University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Sabrina Komatsu
- Human Genome and Stem-Cell Research Center (HUG-CEL), Institute of Biosciences, University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Fernanda Chianca
- Human Genome and Stem-Cell Research Center (HUG-CEL), Institute of Biosciences, University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Luiz Carlos Caires-Júnior
- Human Genome and Stem-Cell Research Center (HUG-CEL), Institute of Biosciences, University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Bruno Henrique Silva Araujo
- Lieber Institute for Brain Development, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Ernesto Goulart
- Human Genome and Stem-Cell Research Center (HUG-CEL), Institute of Biosciences, University of Sao Paulo (USP), Sao Paulo, Brazil
- *Correspondence: Ernesto Goulart, ; Mayana Zatz,
| | - Mayana Zatz
- Human Genome and Stem-Cell Research Center (HUG-CEL), Institute of Biosciences, University of Sao Paulo (USP), Sao Paulo, Brazil
- *Correspondence: Ernesto Goulart, ; Mayana Zatz,
| |
Collapse
|
6
|
Rashidi H, Hay DC. Serum-Free Production of Three-Dimensional Hepatospheres from Pluripotent Stem Cells. Methods Mol Biol 2022; 2454:305-316. [PMID: 34611817 DOI: 10.1007/7651_2021_430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Developing renewable human liver tissue from stem cells has been pursued as a potential source of biological material for pharmaceutical and clinical endeavors. At present, two-dimensional differentiation procedures deliver tissue lacking long-term phenotypic and functional stability. Efforts to overcome these limiting factors have led to the development of protocols to generate three-dimensional cellular aggregates. Here we describe a methodology to generate 3D hepatospheres from human pluripotent stem cells using defined and commercially available reagents.
Collapse
Affiliation(s)
- Hassan Rashidi
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK.
- Institute for Liver and Digestive Health, University College London, London, UK.
| | - David C Hay
- Institute for Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
7
|
Li R, Zhao Y, Yourick JJ, Sprando RL, Gao X. Homogeneous Differentiation of Functional Hepatocytes from Human Induced Pluripotent Stem Cells. Methods Mol Biol 2022; 2429:127-142. [PMID: 35507159 DOI: 10.1007/978-1-0716-1979-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Hepatocyte-like cells (HLCs) generated from human induced pluripotent stem cells (iPSCs) could provide an unlimited source of liver cells for regenerative medicine, disease modeling, drug screening, and toxicology studies. Here we describe a stepwise improved protocol that enables highly efficient, homogeneous, and reproducible differentiation of human iPSCs into functional hepatocytes through controlling all three stages of hepatocyte differentiation, starting from a single cell (non-colony) culture of iPSCs, through homogeneous definitive endoderm induction and highly efficient hepatic specification, and finally arriving at matured HLCs. The final population of cells exhibits morphology closely resembling that of primary human hepatocytes, and expresses specific hepatic markers as evidenced by immunocytochemical staining. More importantly, these HLCs demonstrate key functional characteristics of mature hepatocytes, including major serum protein (e.g., albumin, fibronectin, and alpha-1 antitrypsin) secretion, urea synthesis, glycogen storage, and inducible cytochrome P450 activity.
Collapse
Affiliation(s)
- Rong Li
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, USA
| | - Yang Zhao
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, USA
| | - Jeffrey J Yourick
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, USA
| | - Robert L Sprando
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, USA
| | - Xiugong Gao
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, USA.
| |
Collapse
|
8
|
Pluripotent Stem Cell-Derived Hepatocytes Inhibit T Cell Proliferation In Vitro through Tryptophan Starvation. Cells 2021; 11:cells11010024. [PMID: 35011586 PMCID: PMC8750013 DOI: 10.3390/cells11010024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 11/17/2022] Open
Abstract
Regenerative medicine aims to replace damaged tissues by stimulating endogenous tissue repair or by transplanting autologous or allogeneic cells. Due to their capacity to produce unlimited numbers of cells of a given cell type, pluripotent stem cells, whether of embryonic origin or induced via the reprogramming of somatic cells, are of considerable therapeutic interest in the regenerative medicine field. However, regardless of the cell type, host immune responses present a barrier to success. The aim of this study was to investigate in vitro the immunological properties of human pluripotent stem cell (PSC)-derived hepatocyte-like cells (HLCs). These cells expressed MHC class I molecules while they lacked MHC class II and co-stimulatory molecules, such as CD80 and CD86. Following stimulation with IFN-γ, HLCs upregulated CD40, PD-L1 and MHC class I molecules. When co-cultured with allogeneic T cells, HLCs did not induce T cell proliferation; furthermore, when T cells were stimulated via αCD3/CD28 beads, HLCs inhibited their proliferation via IDO1 and tryptophan deprivation. These results demonstrate that PSC-derived HLCs possess immunoregulatory functions, at least in vitro.
Collapse
|
9
|
Völkner C, Liedtke M, Untucht R, Hermann A, Frech MJ. Patient-Specific iPSC-Derived Neural Differentiated and Hepatocyte-like Cells, Carrying the Compound Heterozygous Mutation p.V1023Sfs*15/p.G992R, Present the "Variant" Biochemical Phenotype of Niemann-Pick Type C1 Disease. Int J Mol Sci 2021; 22:ijms222212184. [PMID: 34830064 PMCID: PMC8624182 DOI: 10.3390/ijms222212184] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 02/07/2023] Open
Abstract
Niemann–Pick disease type C1 (NP-C1) is a rare lysosomal storage disorder caused by autosomal recessive mutations in the NPC1 gene. Patients display a wide spectrum on the clinical as well as on the molecular level, wherein a so-called “variant” biochemical phenotype can be observed. Here, we report an in vitro analysis of fibroblasts obtained from an NP-C1 patient carrying the undescribed compound heterozygous mutation p.V1023Sfs*15/p.G992R. Since NP-C1 is a neurovisceral disease and the patient suffers from severe neurological as well as hepatic symptoms, we extended our study to neural differentiated and hepatocyte-like cells derived from patient-specific induced pluripotent stem cells. We detected slightly increased intracellular cholesterol levels compared to the control cell line in fibroblasts, neural differentiated and hepatocyte-like cells, suggesting a “variant” biochemical phenotype. Furthermore, the total NPC1 protein, as well as post-ER glycoforms of the NPC1 protein, tended to be reduced. In addition, colocalization analysis revealed a mild reduction of the NPC1 protein in the lysosomes. The patient was diagnosed with NP-C1 at the age of 34 years, after an initial misdiagnosis of schizophrenia. After years of mild and unspecific symptoms, such as difficulties in coordination and concentration, symptoms progressed and the patient finally presented with ataxia, dysarthria, dysphagia, vertical supranuclear gaze palsy, and hepatosplenomegaly. Genetic testing finally pointed towards an NP-C1 diagnosis, revealing the so-far undescribed compound heterozygous mutation p.V1023Sfs*15/p.G992R in the NPC1 gene. In light of these findings, this case provides support for the p.G992R mutation being causative for a “variant” biochemical phenotype leading to an adult-onset type of NP-C1 disease.
Collapse
Affiliation(s)
- Christin Völkner
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, 18147 Rostock, Germany; (C.V.); (M.L.); (A.H.)
| | - Maik Liedtke
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, 18147 Rostock, Germany; (C.V.); (M.L.); (A.H.)
| | - Robert Untucht
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany;
| | - Andreas Hermann
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, 18147 Rostock, Germany; (C.V.); (M.L.); (A.H.)
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, 18147 Rostock, Germany
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, 18147 Rostock, Germany
| | - Moritz J. Frech
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, 18147 Rostock, Germany; (C.V.); (M.L.); (A.H.)
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, 18147 Rostock, Germany
- Correspondence:
| |
Collapse
|
10
|
Meseguer-Ripolles J, Lucendo-Villarin B, Tucker C, Ferreira-Gonzalez S, Homer N, Wang Y, Starkey Lewis PJ, M Toledo E, Mellado-Gomez E, Simpson J, Flint O, Jaiswal H, Beer NL, Karlsen AE, Forbes SJ, Dear JW, Hughes J, Hay DC. Dimethyl fumarate reduces hepatocyte senescence following paracetamol exposure. iScience 2021; 24:102552. [PMID: 34151225 PMCID: PMC8188562 DOI: 10.1016/j.isci.2021.102552] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/07/2021] [Accepted: 05/14/2021] [Indexed: 12/30/2022] Open
Abstract
Liver disease is a major cause of premature death. Oxidative stress in the liver represents a key disease driver. Compounds, such as dimethyl fumarate (DMF), can activate the antioxidant response and are used clinically to treat disease. In this study, we tested the protective properties of DMF before or after paracetamol exposure. Following DMF administration, Nrf2 nuclear translocation was tracked at the single-cell level and target gene transactivation confirmed. Next, the protective properties of DMF were examined following paracetamol exposure. Transcriptomic and biochemical analysis revealed that DMF rescue was underpinned by reduced Nf-kB and TGF-β signaling and cell senescence. Following on from these studies, we employed a Zebrafish model to study paracetamol exposure in vivo. We combined a genetically modified Zebrafish model, expressing green fluorescent protein exclusively in the liver, with automated microscopy. Pre-treatment with DMF, prior to paracetamol exposure, led to reduced liver damage in Zebrafish demonstrating protective properties.
Collapse
Affiliation(s)
- Jose Meseguer-Ripolles
- Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Baltasar Lucendo-Villarin
- Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Carl Tucker
- Centre for Inflammation Research, University of Edinburgh, 47 Little France Drive, Edinburgh EH16 4TJ, UK
| | - Sofia Ferreira-Gonzalez
- Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Natalie Homer
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Drive, Edinburgh, UK
| | - Yu Wang
- Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Philip J. Starkey Lewis
- Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Enrique M Toledo
- Novo Nordisk Research Centre Oxford (NNRCO), Novo Nordisk Ltd, Innovation Building - Old Road Campus Roosevelt Drive, OX3 7FZ Oxford, UK
- Novo Nordisk Ltd, Novo Nordisk Park 1, 2760 Måløv, Denmark
| | - Esther Mellado-Gomez
- Novo Nordisk Research Centre Oxford (NNRCO), Novo Nordisk Ltd, Innovation Building - Old Road Campus Roosevelt Drive, OX3 7FZ Oxford, UK
- Novo Nordisk Ltd, Novo Nordisk Park 1, 2760 Måløv, Denmark
| | - Joanna Simpson
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Drive, Edinburgh, UK
| | - Oliver Flint
- Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Himjyot Jaiswal
- Novo Nordisk Research Centre Oxford (NNRCO), Novo Nordisk Ltd, Innovation Building - Old Road Campus Roosevelt Drive, OX3 7FZ Oxford, UK
- Novo Nordisk Ltd, Novo Nordisk Park 1, 2760 Måløv, Denmark
| | - Nicola L. Beer
- Novo Nordisk Research Centre Oxford (NNRCO), Novo Nordisk Ltd, Innovation Building - Old Road Campus Roosevelt Drive, OX3 7FZ Oxford, UK
- Novo Nordisk Ltd, Novo Nordisk Park 1, 2760 Måløv, Denmark
| | - Allan E. Karlsen
- Novo Nordisk Research Centre Oxford (NNRCO), Novo Nordisk Ltd, Innovation Building - Old Road Campus Roosevelt Drive, OX3 7FZ Oxford, UK
- Novo Nordisk Ltd, Novo Nordisk Park 1, 2760 Måløv, Denmark
| | - Stuart J. Forbes
- Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - James W. Dear
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Drive, Edinburgh, UK
| | - Jeremy Hughes
- Centre for Inflammation Research, University of Edinburgh, 47 Little France Drive, Edinburgh EH16 4TJ, UK
| | - David C. Hay
- Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| |
Collapse
|
11
|
Sinton MC, Meseguer-Ripolles J, Lucendo-Villarin B, Drake AJ, Hay DC. Modeling human hepatic steatosis in pluripotent stem cell-derived hepatocytes. STAR Protoc 2021; 2:100493. [PMID: 33997813 PMCID: PMC8091923 DOI: 10.1016/j.xpro.2021.100493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
This protocol describes the production of hepatocyte-like cells (HLCs) from human pluripotent stem cells and how to induce hepatic steatosis, a condition characterized by intracellular lipid accumulation. Following differentiation to an HLC phenotype, intracellular lipid accumulation is induced with a steatosis induction cocktail, allowing the user to examine the cellular processes that underpin hepatic steatosis. Furthermore, the renewable nature of our system, on a defined genetic background, permits in-depth mechanistic analysis, which may facilitate therapeutic target identification in the future. For complete details on the use and execution of this protocol, please refer to Sinton et al. (2021).
Collapse
Affiliation(s)
- Matthew C. Sinton
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Jose Meseguer-Ripolles
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, 5 Little France Crescent, Edinburgh EH16 4UU, UK
| | - Baltasar Lucendo-Villarin
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, 5 Little France Crescent, Edinburgh EH16 4UU, UK
| | - Amanda J. Drake
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - David C. Hay
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter, 5 Little France Crescent, Edinburgh EH16 4UU, UK
| |
Collapse
|
12
|
Meseguer-Ripolles J, Kasarinaite A, Lucendo-Villarin B, Hay DC. Protocol for automated production of human stem cell derived liver spheres. STAR Protoc 2021; 2:100502. [PMID: 33997816 PMCID: PMC8105683 DOI: 10.1016/j.xpro.2021.100502] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This protocol describes how to produce human liver spheres from pluripotent stem cell-derived hepatic progenitors, endothelial cells, and hepatic stellate cells. Liver spheres form by self-assembly in microwells, generating up to 73 spheres per well of a 96-well plate. This process was automated using liquid handling and pipetting systems, permitting cost-effective scale-up and reducing sphere variability. In its current form, this system provides a powerful tool to generate human liver tissue for disease modeling and drug screening. For complete details on the use and execution of this protocol, please refer to Lucendo-Villarin et al. (2020) (https://doi.org/10.1088/1758-5090/abbdb2). Protocol to produce and self-assemble hepatocytes, endothelial, and stellate cells Generation of an automated system for 3D liver sphere production Scalable production of phenotypically stable liver spheres for disease modeling
Collapse
Affiliation(s)
- Jose Meseguer-Ripolles
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Alvile Kasarinaite
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Baltasar Lucendo-Villarin
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - David C Hay
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK
| |
Collapse
|
13
|
Leedale JA, Lucendo-Villarin B, Meseguer-Ripolles J, Kasarinaite A, Webb SD, Hay DC. Mathematical modelling of oxygen gradients in stem cell-derived liver tissue. PLoS One 2021; 16:e0244070. [PMID: 33556073 PMCID: PMC7870006 DOI: 10.1371/journal.pone.0244070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/03/2020] [Indexed: 01/08/2023] Open
Abstract
A major bottleneck in the study of human liver physiology is the provision of stable liver tissue in sufficient quantity. As a result, current approaches to modelling human drug efficacy and toxicity rely heavily on immortalized human and animal cell lines. These models are informative but do possess significant drawbacks. To address the issues presented by those models, researchers have turned to pluripotent stem cells (PSCs). PSCs can be generated from defined genetic backgrounds, are scalable, and capable of differentiation to all the cell types found in the human body, representing an attractive source of somatic cells for in vitro and in vivo endeavours. Although unlimited numbers of somatic cell types can be generated in vitro, their maturation still remains problematic. In order to develop high fidelity PSC-derived liver tissue, it is necessary to better understand the cell microenvironment in vitro including key elements of liver physiology. In vivo a major driver of zonated liver function is the oxygen gradient that exists from periportal to pericentral regions. In this paper, we demonstrate how cell culture conditions for PSC-derived liver sphere systems can be optimised to recapitulate physiologically relevant oxygen gradients by using mathematical modelling. The mathematical model incorporates some often-understated features and mechanisms of traditional spheroid systems such as cell-specific oxygen uptake, media volume, spheroid size, and well dimensions that can lead to a spatially heterogeneous distribution of oxygen. This mathematical modelling approach allows for the calibration and identification of culture conditions required to generate physiologically realistic function within the microtissue through recapitulation of the in vivo microenvironment.
Collapse
Affiliation(s)
- Joseph A. Leedale
- Department of Mathematical Sciences, University of Liverpool, Liverpool, United Kingdom
- * E-mail: (JAL); (DCH)
| | | | - Jose Meseguer-Ripolles
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Alvile Kasarinaite
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Steven D. Webb
- Department of Applied Mathematics, Liverpool John Moores University, Liverpool, United Kingdom
| | - David C. Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
- * E-mail: (JAL); (DCH)
| |
Collapse
|
14
|
Sinton MC, Meseguer-Ripolles J, Lucendo-Villarin B, Wernig-Zorc S, Thomson JP, Carter RN, Lyall MJ, Walker PD, Thakker A, Meehan RR, Lavery GG, Morton NM, Ludwig C, Tennant DA, Hay DC, Drake AJ. A human pluripotent stem cell model for the analysis of metabolic dysfunction in hepatic steatosis. iScience 2021; 24:101931. [PMID: 33409477 PMCID: PMC7773967 DOI: 10.1016/j.isci.2020.101931] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/20/2020] [Accepted: 12/08/2020] [Indexed: 02/08/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is currently the most prevalent form of liver disease worldwide. This term encompasses a spectrum of pathologies, from benign hepatic steatosis to non-alcoholic steatohepatitis, which have, to date, been challenging to model in the laboratory setting. Here, we present a human pluripotent stem cell (hPSC)-derived model of hepatic steatosis, which overcomes inherent challenges of current models and provides insights into the metabolic rewiring associated with steatosis. Following induction of macrovesicular steatosis in hepatocyte-like cells using lactate, pyruvate, and octanoate (LPO), respirometry and transcriptomic analyses revealed compromised electron transport chain activity. 13C isotopic tracing studies revealed enhanced TCA cycle anaplerosis, with concomitant development of a compensatory purine nucleotide cycle shunt leading to excess generation of fumarate. This model of hepatic steatosis is reproducible, scalable, and overcomes the challenges of studying mitochondrial metabolism in currently available models.
Collapse
Affiliation(s)
- Matthew C. Sinton
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Jose Meseguer-Ripolles
- Centre for Regenerative Medicine, University of Edinburgh, Institute for Regeneration and Repair, Edinburgh BioQuarter, 5 Little France Crescent, Edinburgh, EH16 4UU, UK
| | - Baltasar Lucendo-Villarin
- Centre for Regenerative Medicine, University of Edinburgh, Institute for Regeneration and Repair, Edinburgh BioQuarter, 5 Little France Crescent, Edinburgh, EH16 4UU, UK
| | - Sara Wernig-Zorc
- Department of Biochemistry, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - John P. Thomson
- Human Genetics Unit, University of Edinburgh, MRC Institute for Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4, 2XU, UK
| | - Roderick N. Carter
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Marcus J. Lyall
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Paul D. Walker
- Institute of Metabolism and Systems Research, IBR Tower, College of Medical and Dental Sciences, Edgbaston, University of Birmingham, Birmingham, B15 2TT,, UK
| | - Alpesh Thakker
- Institute of Metabolism and Systems Research, IBR Tower, College of Medical and Dental Sciences, Edgbaston, University of Birmingham, Birmingham, B15 2TT,, UK
| | - Richard R. Meehan
- Human Genetics Unit, University of Edinburgh, MRC Institute for Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4, 2XU, UK
| | - Gareth G. Lavery
- Institute of Metabolism and Systems Research, IBR Tower, College of Medical and Dental Sciences, Edgbaston, University of Birmingham, Birmingham, B15 2TT,, UK
| | - Nicholas M. Morton
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Christian Ludwig
- Institute of Metabolism and Systems Research, IBR Tower, College of Medical and Dental Sciences, Edgbaston, University of Birmingham, Birmingham, B15 2TT,, UK
| | - Daniel A. Tennant
- Institute of Metabolism and Systems Research, IBR Tower, College of Medical and Dental Sciences, Edgbaston, University of Birmingham, Birmingham, B15 2TT,, UK
| | - David C. Hay
- Centre for Regenerative Medicine, University of Edinburgh, Institute for Regeneration and Repair, Edinburgh BioQuarter, 5 Little France Crescent, Edinburgh, EH16 4UU, UK
| | - Amanda J. Drake
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
15
|
Cox CR, Lynch S, Goldring C, Sharma P. Current Perspective: 3D Spheroid Models Utilizing Human-Based Cells for Investigating Metabolism-Dependent Drug-Induced Liver Injury. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:611913. [PMID: 35047893 PMCID: PMC8757888 DOI: 10.3389/fmedt.2020.611913] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
Drug-induced liver injury (DILI) remains a leading cause for the withdrawal of approved drugs. This has significant financial implications for pharmaceutical companies, places increasing strain on global health services, and causes harm to patients. For these reasons, it is essential that in-vitro liver models are capable of detecting DILI-positive compounds and their underlying mechanisms, prior to their approval and administration to patients or volunteers in clinical trials. Metabolism-dependent DILI is an important mechanism of drug-induced toxicity, which often involves the CYP450 family of enzymes, and is associated with the production of a chemically reactive metabolite and/or inefficient removal and accumulation of potentially toxic compounds. Unfortunately, many of the traditional in-vitro liver models fall short of their in-vivo counterparts, failing to recapitulate the mature hepatocyte phenotype, becoming metabolically incompetent, and lacking the longevity to investigate and detect metabolism-dependent DILI and those associated with chronic and repeat dosing regimens. Nevertheless, evidence is gathering to indicate that growing cells in 3D formats can increase the complexity of these models, promoting a more mature-hepatocyte phenotype and increasing their longevity, in vitro. This review will discuss the use of 3D in vitro models, namely spheroids, organoids, and perfusion-based systems to establish suitable liver models to investigate metabolism-dependent DILI.
Collapse
Affiliation(s)
- Christopher R. Cox
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- *Correspondence: Christopher R. Cox
| | - Stephen Lynch
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Christopher Goldring
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Parveen Sharma
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Liverpool Centre for Cardiovascular Science, Liverpool, United Kingdom
| |
Collapse
|
16
|
Lucendo-Villarin B, Nell P, Hellwig B, Filis P, Feuerborn D, O'Shaughnessy PJ, Godoy P, Rahnenführer J, Hengstler JG, Cherianidou A, Sachinidis A, Fowler PA, Hay DC. Genome-wide expression changes induced by bisphenol A, F and S in human stem cell derived hepatocyte-like cells. EXCLI JOURNAL 2020; 19:1459-1476. [PMID: 33312107 PMCID: PMC7726493 DOI: 10.17179/excli2020-2934] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022]
Abstract
The debate about possible adverse effects of bisphenol A (BPA) has been ongoing for decades. Bisphenol F (BPF) and S (BPS) have been suggested as “safer” alternatives. In the present study we used hepatocyte-like cells (HLCs) derived from the human embryonic stem cell lines Man12 and H9 to compare the three bisphenol derivatives. Stem cell-derived progenitors were produced using an established system and were exposed to BPA, BPF and BPS for 8 days during their transition to HLCs. Subsequently, we examined cell viability, inhibition of cytochrome P450 (CYP) activity, and genome-wide RNA profiles. Sub-cytotoxic, inhibitory concentrations (IC50) of CYP3A were 20, 9.5 and 25 µM for BPA, BPF and BPS in Man12 derived HLCs, respectively. The corresponding concentrations for H9-derived HLCs were 19, 29 and 31 µM. These IC50 concentrations were used to study global expression changes in this in vitro study and are higher than unconjugated BPA in serum of the general population. A large overlap of up- as well as downregulated genes induced by the three bisphenol derivatives was seen. This is at least 28-fold higher compared to randomly expected gene expression changes. Moreover, highly significant correlations of expression changes induced by the three bisphenol derivatives were obtained in pairwise comparisons. Dysregulated genes were associated with reduced metabolic function, cellular differentiation, embryonic development, cell survival and apoptosis. In conclusion, no major differences in cytochrome inhibitory activities of BPA, BPF and BPS were observed and gene expression changes showed a high degree of similarity.
Collapse
Affiliation(s)
- B Lucendo-Villarin
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - P Nell
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - B Hellwig
- Department of Statistics, Technical University Dortmund, Dortmund, Germany
| | - P Filis
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - D Feuerborn
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - P J O'Shaughnessy
- Institute of Biodiversity, Animal Health & Comparative Medicine, University of Glasgow, UK
| | - P Godoy
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - J Rahnenführer
- Department of Statistics, Technical University Dortmund, Dortmund, Germany
| | - J G Hengstler
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - A Cherianidou
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Cologne, Germany
| | - A Sachinidis
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Cologne, Germany
| | - P A Fowler
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - D C Hay
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
17
|
Lucendo-Villarin B, Meseguer-Ripolles J, Drew J, Fischer L, Ma E, Flint O, Simpson KJ, Machesky LM, Mountford JC, Hay DC. Development of a cost-effective automated platform to produce human liver spheroids for basic and applied research. Biofabrication 2020; 13:015009. [PMID: 33007774 DOI: 10.1088/1758-5090/abbdb2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/02/2020] [Indexed: 12/14/2022]
Abstract
Liver disease represents an increasing cause of global morbidity and mortality. Currently, liver transplant is the only treatment curative for end-stage liver disease. Donor organs cannot meet the demand and therefore scalable treatments and new disease models are required to improve clinical intervention. Pluripotent stem cells represent a renewable source of human tissue. Recent advances in three-dimensional cell culture have provided the field with more complex systems that better mimic liver physiology and function. Despite these improvements, current cell-based models are variable in performance and expensive to manufacture at scale. This is due, in part, to the use of poorly defined or cross-species materials within the process, severely affecting technology translation. To address this issue, we have developed an automated and economical platform to produce liver tissue at scale for modelling disease and small molecule screening. Stem cell derived liver spheres were formed by combining hepatic progenitors with endothelial cells and stellate cells, in the ratios found within the liver. The resulting tissue permitted the study of human liver biology 'in the dish' and could be scaled for screening. In summary, we have developed an automated differentiation system that permits reliable self-assembly of human liver tissue for biomedical application. Going forward we believe that this technology will not only serve as anin vitroresource, and may have an important role to play in supporting failing liver function in humans.
Collapse
Affiliation(s)
- B Lucendo-Villarin
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
- Both authors contributed equally to this manuscript
| | - J Meseguer-Ripolles
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
- Both authors contributed equally to this manuscript
| | - J Drew
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, United Kingdom
| | - L Fischer
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - E Ma
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Campus, G61 1BD, United Kingdom
| | - O Flint
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - K J Simpson
- Scottish Liver Transplant Unit, Royal Infirmary, Edinburgh EH16 4SA, United Kingdom
| | - L M Machesky
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Campus, G61 1BD, United Kingdom
| | - J C Mountford
- SNBTS, 52 Research Avenue North, Heriot-Watt Research Park, Edinburgh EH14 4BE, United Kingdom
| | - D C Hay
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
- Author to whom any correspondence should be addressed
| |
Collapse
|
18
|
Fischer L, Lucendo-Villarin B, Hay DC, O’Farrelly C. Human PSC-Derived Hepatocytes Express Low Levels of Viral Pathogen Recognition Receptors, but Are Capable of Mounting an Effective Innate Immune Response. Int J Mol Sci 2020; 21:ijms21113831. [PMID: 32481600 PMCID: PMC7312201 DOI: 10.3390/ijms21113831] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 04/30/2020] [Accepted: 05/24/2020] [Indexed: 01/19/2023] Open
Abstract
Hepatocytes are key players in the innate immune response to liver pathogens but are challenging to study because of inaccessibility and a short half-life. Recent advances in in vitro differentiation of hepatocyte-like cells (HLCs) facilitated studies of hepatocyte-pathogen interactions. Here, we aimed to define the anti-viral innate immune potential of human HLCs with a focus on toll-like receptor (TLR)-expression and the presence of a metabolic switch. We analysed cytoplasmic pattern recognition receptor (PRR)- and endosomal TLR-expression and activity and adaptation of HLCs to an inflammatory environment. We found that transcript levels of retinoic acid inducible gene I (RIG-I), melanoma differentiation antigen 5 (MDA5), and TLR3 became downregulated during differentiation, indicating the acquisition of a more tolerogenic phenotype, as expected in healthy hepatocytes. HLCs responded to activation of RIG-I by producing interferons (IFNs) and IFN-stimulated genes. Despite low-level expression of TLR3, receptor expression was upregulated in an inflammatory environment. TLR3 signalling induced expression of proinflammatory cytokines at the gene level, indicating that several PRRs need to interact for successful innate immune activation. The inflammatory responsiveness of HLCs was accompanied by the downregulation of cytochrome P450 3A and 1A2 activity and decreased serum protein production, showing that the metabolic switch seen in primary hepatocytes during anti-viral responses is also present in HLCs.
Collapse
Affiliation(s)
- Lena Fischer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland;
| | | | - David C. Hay
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK;
- Correspondence: (D.C.H.); (C.O.)
| | - Cliona O’Farrelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland;
- School of Medicine, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin 2, Ireland
- Correspondence: (D.C.H.); (C.O.)
| |
Collapse
|
19
|
Goulart E, de Caires-Junior LC, Telles-Silva KA, Araujo BHS, Rocco SA, Sforca M, de Sousa IL, Kobayashi GS, Musso CM, Assoni AF, Oliveira D, Caldini E, Raia S, Lelkes PI, Zatz M. 3D bioprinting of liver spheroids derived from human induced pluripotent stem cells sustain liver function and viability
in vitro. Biofabrication 2019; 12:015010. [DOI: 10.1088/1758-5090/ab4a30] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
20
|
Generation of qualified clinical-grade functional hepatocytes from human embryonic stem cells in chemically defined conditions. Cell Death Dis 2019; 10:763. [PMID: 31601782 PMCID: PMC6787193 DOI: 10.1038/s41419-019-1967-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 08/10/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022]
Abstract
Hepatocytes have been successfully generated from human pluripotent stem cells (hPSCs). However, the cost-effective and clinical-grade generation of hepatocytes from hPSCs still need to be improved. In this study, we reported the production of functional hepatocytes from clinical-grade human embryonic stem cells (hESCs) under good manufacturing practice (GMP) requirements. We sequentially generated primitive streak (PS), definitive endoderm (DE), hepatoblasts and hepatocyte-like cells (HLCs) from hESCs in the different stages with completely defined reagents. During hepatoblast differentiation, dimethyl sulfoxide (DMSO), transferrin, L-ascorbic acid 2-phosphate sesquimagnesium salt hydrate (Vc-Mg), insulin, and sodium selenite were used instead of cytokines and FBS/KOSR. Then, hepatoblasts were differentiated into HLCs that had a typical hepatocyte morphology and possessed characteristics of mature hepatocytes, such as metabolic-related gene expression, albumin secretion, fat accumulation, glycogen storage, and inducible cytochrome P450 activity in vitro. HLCs integrated into the livers of Tet-uPA Rag2–/– Il2rg–/– (URG) mice, which partially recovered after transplantation. Furthermore, a series of biosafety-related experiments were performed to ensure future clinical applications. In conclusion, we developed a chemically defined system to generate qualified clinical-grade HLCs from hESCs under GMP conditions. HLCs have been proven to be safe and effective for treating liver failure. This efficient platform could facilitate the treatment of liver diseases using hESC-derived HLCs transplantation.
Collapse
|
21
|
Wang Y, Tatham MH, Schmidt-Heck W, Swann C, Singh-Dolt K, Meseguer-Ripolles J, Lucendo-Villarin B, Kunath T, Rudd TR, Smith AJH, Hengstler JG, Godoy P, Hay RT, Hay DC. Multiomics Analyses of HNF4α Protein Domain Function during Human Pluripotent Stem Cell Differentiation. iScience 2019; 16:206-217. [PMID: 31185456 PMCID: PMC6556878 DOI: 10.1016/j.isci.2019.05.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/08/2019] [Accepted: 05/21/2019] [Indexed: 02/06/2023] Open
Abstract
During mammalian development, liver differentiation is driven by signals that converge on multiple transcription factor networks. The hepatocyte nuclear factor signaling network is known to be essential for hepatocyte specification and maintenance. In this study, we have generated deletion and point mutants of hepatocyte nuclear factor-4alpha (HNF4α) to precisely evaluate the function of protein domains during hepatocyte specification from human pluripotent stem cells. We demonstrate that nuclear HNF4α is essential for hepatic progenitor specification, and the introduction of point mutations in HNF4α's Small Ubiquitin-like Modifier (SUMO) consensus motif leads to disrupted hepatocyte differentiation. Taking a multiomics approach, we identified key deficiencies in cell biology, which included dysfunctional metabolism, substrate adhesion, tricarboxylic acid cycle flux, microRNA transport, and mRNA processing. In summary, the combination of genome editing and multiomics analyses has provided valuable insight into the diverse functions of HNF4α during pluripotent stem cell entry into the hepatic lineage and during hepatocellular differentiation.
Collapse
Affiliation(s)
- Yu Wang
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, Scotland EH16 4UU, UK
| | - Michael H Tatham
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Wolfgang Schmidt-Heck
- Leibniz Institute for Natural Product Research and Infection Biology eV-Hans-Knoll Institute, Jena, Germany
| | - Carolyn Swann
- National Institute for Biological Standards and Control (MHRA), Blanche Lane, South Mimms, Hertfordshire EN6 3QG, UK
| | - Karamjit Singh-Dolt
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, Scotland EH16 4UU, UK
| | - Jose Meseguer-Ripolles
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, Scotland EH16 4UU, UK
| | - Baltasar Lucendo-Villarin
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, Scotland EH16 4UU, UK
| | - Tilo Kunath
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, Scotland EH16 4UU, UK
| | - Timothy R Rudd
- National Institute for Biological Standards and Control (MHRA), Blanche Lane, South Mimms, Hertfordshire EN6 3QG, UK
| | - Andrew J H Smith
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, Scotland EH16 4UU, UK
| | - Jan G Hengstler
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - Patricio Godoy
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - Ronald T Hay
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - David C Hay
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, Scotland EH16 4UU, UK.
| |
Collapse
|
22
|
Natale A, Vanmol K, Arslan A, Van Vlierberghe S, Dubruel P, Van Erps J, Thienpont H, Buzgo M, Boeckmans J, De Kock J, Vanhaecke T, Rogiers V, Rodrigues RM. Technological advancements for the development of stem cell-based models for hepatotoxicity testing. Arch Toxicol 2019; 93:1789-1805. [PMID: 31037322 DOI: 10.1007/s00204-019-02465-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/18/2019] [Indexed: 02/07/2023]
Abstract
Stem cells are characterized by their self-renewal capacity and their ability to differentiate into multiple cell types of the human body. Using directed differentiation strategies, stem cells can now be converted into hepatocyte-like cells (HLCs) and therefore, represent a unique cell source for toxicological applications in vitro. However, the acquired hepatic functionality of stem cell-derived HLCs is still significantly inferior to primary human hepatocytes. One of the main reasons for this is that most in vitro models use traditional two-dimensional (2D) setups where the flat substrata cannot properly mimic the physiology of the human liver. Therefore, 2D-setups are progressively being replaced by more advanced culture systems, which attempt to replicate the natural liver microenvironment, in which stem cells can better differentiate towards HLCs. This review highlights the most recent cell culture systems, including scaffold-free and scaffold-based three-dimensional (3D) technologies and microfluidics that can be employed for culture and hepatic differentiation of stem cells intended for hepatotoxicity testing. These methodologies have shown to improve in vitro liver cell functionality according to the in vivo liver physiology and allow to establish stem cell-based hepatic in vitro platforms for the accurate evaluation of xenobiotics.
Collapse
Affiliation(s)
- Alessandra Natale
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Koen Vanmol
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
| | - Aysu Arslan
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Sandra Van Vlierberghe
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Peter Dubruel
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Jürgen Van Erps
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
| | - Hugo Thienpont
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
| | | | - Joost Boeckmans
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Joery De Kock
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Tamara Vanhaecke
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Vera Rogiers
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Robim M Rodrigues
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
23
|
Iansante V, Chandrashekran A, Dhawan A. Cell-based liver therapies: past, present and future. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0229. [PMID: 29786563 DOI: 10.1098/rstb.2017.0229] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2017] [Indexed: 12/16/2022] Open
Abstract
Liver transplantation represents the standard treatment for people with an end-stage liver disease and some liver-based metabolic disorders; however, shortage of liver donor tissues limits its availability. Furthermore, whole liver replacement eliminates the possibility of using native liver as a possible target for future gene therapy in case of liver-based metabolic defects. Cell therapy has emerged as a potential alternative, as cells can provide the hepatic functions and engraft in the liver parenchyma. Various options have been proposed, including human or other species hepatocytes, hepatocyte-like cells derived from stem cells or more futuristic alternatives, such as combination therapies with different cell types, organoids and cell-biomaterial combinations. In this review, we aim to give an overview of the cell therapies developed so far, highlighting preclinical and/or clinical achievements as well as the limitations that need to be overcome to make them fully effective and safe for clinical applications.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- Valeria Iansante
- Dhawan Lab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London at King's College Hospital, London SE5 9PJ, UK
| | - Anil Chandrashekran
- Dhawan Lab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London at King's College Hospital, London SE5 9PJ, UK
| | - Anil Dhawan
- Dhawan Lab, Paediatric Liver GI and Nutrition Center and MowatLabs, Institute of Liver Studies, King's College London at King's College Hospital, London SE5 9PJ, UK
| |
Collapse
|
24
|
Rashidi H, Luu NT, Alwahsh SM, Ginai M, Alhaque S, Dong H, Tomaz RA, Vernay B, Vigneswara V, Hallett JM, Chandrashekran A, Dhawan A, Vallier L, Bradley M, Callanan A, Forbes SJ, Newsome PN, Hay DC. 3D human liver tissue from pluripotent stem cells displays stable phenotype in vitro and supports compromised liver function in vivo. Arch Toxicol 2018; 92:3117-3129. [PMID: 30155720 PMCID: PMC6132688 DOI: 10.1007/s00204-018-2280-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 07/31/2018] [Indexed: 12/15/2022]
Abstract
Liver disease is an escalating global health issue. While liver transplantation is an effective mode of therapy, patient mortality has increased due to the shortage of donor organs. Developing renewable sources of human liver tissue is therefore attractive. Pluripotent stem cell-derived liver tissue represents a potential alternative to cadaver derived hepatocytes and whole organ transplant. At present, two-dimensional differentiation procedures deliver tissue lacking certain functions and long-term stability. Efforts to overcome these limiting factors have led to the building of three-dimensional (3D) cellular aggregates. Although enabling for the field, their widespread application is limited due to their reliance on variable biological components. Our studies focused on the development of 3D liver tissue under defined conditions. In vitro generated 3D tissues exhibited stable phenotype for over 1 year in culture, providing an attractive resource for long-term in vitro studies. Moreover, 3D derived tissue provided critical liver support in two animal models, including immunocompetent recipients. Therefore, we believe that our study provides stable human tissue to better model liver biology 'in the dish', and in the future may permit the support of compromised liver function in humans.
Collapse
Affiliation(s)
- Hassan Rashidi
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Nguyet-Thin Luu
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute for Health Research Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, UK
| | - Salamah M Alwahsh
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Maaria Ginai
- Institute of Bioengineering, The University of Edinburgh, King's Buildings, Edinburgh, EH9 3DW, UK
| | - Sharmin Alhaque
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Hua Dong
- School of Chemistry, University of Edinburgh, Kings Buildings, EH9 3FJ, Edinburgh, UK
| | - Rute A Tomaz
- Anne McLaren Laboratory, Wellcome Trust-MRC Stem Cell Institute, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Bertrand Vernay
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Vasanthy Vigneswara
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute for Health Research Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, UK
| | - John M Hallett
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Anil Chandrashekran
- Child Health Clinical Academic Group, MRC Centre for Transplantation, King's College London, London, UK
| | - Anil Dhawan
- Child Health Clinical Academic Group, MRC Centre for Transplantation, King's College London, London, UK
| | - Ludovic Vallier
- Anne McLaren Laboratory, Wellcome Trust-MRC Stem Cell Institute, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Mark Bradley
- School of Chemistry, University of Edinburgh, Kings Buildings, EH9 3FJ, Edinburgh, UK
| | - Anthony Callanan
- Institute of Bioengineering, The University of Edinburgh, King's Buildings, Edinburgh, EH9 3DW, UK
| | - Stuart J Forbes
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Philip N Newsome
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute for Health Research Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, UK.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - David C Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
25
|
Meseguer-Ripolles J, Lucendo-Villarin B, Wang Y, Hay DC. Semi-automated Production of Hepatocyte Like Cells from Pluripotent Stem Cells. J Vis Exp 2018:57995. [PMID: 30102283 PMCID: PMC6126566 DOI: 10.3791/57995] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human pluripotent stem cells represent a renewable source of human tissue. Our research is focused on generating human liver tissue from human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs). Current differentiation procedures generate human hepatocyte-like cells (HLCs) displaying a mixture of fetal and adult traits. To improve cell phenotype, we have fully defined our differentiation procedure and the cell niche, resulting in the generation of cell populations which display improved gene expression and function. While these studies mark progress, the ability to generate large quantities of multi well plates for screening has been limited by labour intensive procedures and batch to batch variation. To tackle this issue, we have developed a semi-automated platform to differentiate pluripotent stem cells into HLCs. Stem cell seeding and differentiation were performed using liquid handling and automatic pipetting systems in 96-well plate format. Following the differentiation, cell phenotype was analyzed using automated microscopy and a multi well luminometer.
Collapse
Affiliation(s)
| | | | - Yu Wang
- MRC Centre for Regenerative Medicine, University of Edinburgh
| | - David C Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh;
| |
Collapse
|
26
|
Alwahsh SM, Rashidi H, Hay DC. Liver cell therapy: is this the end of the beginning? Cell Mol Life Sci 2018; 75:1307-1324. [PMID: 29181772 PMCID: PMC5852182 DOI: 10.1007/s00018-017-2713-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 11/08/2017] [Accepted: 11/13/2017] [Indexed: 12/13/2022]
Abstract
The prevalence of liver diseases is increasing globally. Orthotopic liver transplantation is widely used to treat liver disease upon organ failure. The complexity of this procedure and finite numbers of healthy organ donors have prompted research into alternative therapeutic options to treat liver disease. This includes the transplantation of liver cells to promote regeneration. While successful, the routine supply of good quality human liver cells is limited. Therefore, renewable and scalable sources of these cells are sought. Liver progenitor and pluripotent stem cells offer potential cell sources that could be used clinically. This review discusses recent approaches in liver cell transplantation and requirements to improve the process, with the ultimate goal being efficient organ regeneration. We also discuss the potential off-target effects of cell-based therapies, and the advantages and drawbacks of current pre-clinical animal models used to study organ senescence, repopulation and regeneration.
Collapse
Affiliation(s)
- Salamah M Alwahsh
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| | - Hassan Rashidi
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - David C Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
27
|
Meseguer-Ripolles J, Khetani SR, Blanco JG, Iredale M, Hay DC. Pluripotent Stem Cell-Derived Human Tissue: Platforms to Evaluate Drug Metabolism and Safety. AAPS J 2017; 20:20. [PMID: 29270863 PMCID: PMC5804345 DOI: 10.1208/s12248-017-0171-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/16/2017] [Indexed: 12/15/2022] Open
Abstract
Despite the improvements in drug screening, high levels of drug attrition persist. Although high-throughput screening platforms permit the testing of compound libraries, poor compound efficacy or unexpected organ toxicity are major causes of attrition. Part of the reason for drug failure resides in the models employed, most of which are not representative of normal organ biology. This same problem affects all the major organs during drug development. Hepatotoxicity and cardiotoxicity are two interesting examples of organ disease and can present in the late stages of drug development, resulting in major cost and increased risk to the patient. Currently, cell-based systems used within industry rely on immortalized or primary cell lines from donated tissue. These models possess significant advantages and disadvantages, but in general display limited relevance to the organ of interest. Recently, stem cell technology has shown promise in drug development and has been proposed as an alternative to current industrial systems. These offerings will provide the field with exciting new models to study human organ biology at scale and in detail. We believe that the recent advances in production of stem cell-derived hepatocytes and cardiomyocytes combined with cutting-edge engineering technologies make them an attractive alternative to current screening models for drug discovery. This will lead to fast failing of poor drugs earlier in the process, delivering safer and more efficacious medicines for the patient.
Collapse
Affiliation(s)
| | - Salman R Khetani
- University of Illinois at Chicago, Bioengineering (MC 063) 851 S Morgan St, 218 SEO, Chicago, Illinois, 60607, USA
| | - Javier G Blanco
- School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Mairi Iredale
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - David C Hay
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|