151
|
Godoy P, Schmidt-Heck W, Natarajan K, Lucendo-Villarin B, Szkolnicka D, Asplund A, Björquist P, Widera A, Stöber R, Campos G, Hammad S, Sachinidis A, Chaudhari U, Damm G, Weiss TS, Nüssler A, Synnergren J, Edlund K, Küppers-Munther B, Hay DC, Hengstler JG. Gene networks and transcription factor motifs defining the differentiation of stem cells into hepatocyte-like cells. J Hepatol 2015; 63:934-942. [PMID: 26022688 PMCID: PMC4580233 DOI: 10.1016/j.jhep.2015.05.013] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 04/25/2015] [Accepted: 05/11/2015] [Indexed: 01/10/2023]
Abstract
BACKGROUND & AIMS The differentiation of stem cells to hepatocyte-like cells (HLC) offers the perspective of unlimited supply of human hepatocytes. However, the degree of differentiation of HLC remains controversial. To obtain an unbiased characterization, we performed a transcriptomic study with HLC derived from human embryonic and induced stem cells (ESC, hiPSC) from three different laboratories. METHODS Genome-wide gene expression profiles of ESC and HLC were compared to freshly isolated and up to 14days cultivated primary human hepatocytes. Gene networks representing successful and failed hepatocyte differentiation, and the transcription factors involved in their regulation were identified. RESULTS Gene regulatory network analysis demonstrated that HLC represent a mixed cell type with features of liver, intestine, fibroblast and stem cells. The "unwanted" intestinal features were associated with KLF5 and CDX2 transcriptional networks. Cluster analysis identified highly correlated groups of genes associated with mature liver functions (n=1057) and downregulated proliferation associated genes (n=1562) that approach levels of primary hepatocytes. However, three further clusters containing 447, 101, and 505 genes failed to reach levels of hepatocytes. Key TF of two of these clusters include SOX11, FOXQ1, and YBX3. The third unsuccessful cluster, controlled by HNF1, CAR, FXR, and PXR, strongly overlaps with genes repressed in cultivated hepatocytes compared to freshly isolated hepatocytes, suggesting that current in vitro conditions lack stimuli required to maintain gene expression in hepatocytes, which consequently also explains a corresponding deficiency of HLC. CONCLUSIONS The present gene regulatory network approach identifies key transcription factors which require modulation to improve HLC differentiation.
Collapse
Affiliation(s)
- Patricio Godoy
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany; Department of Physiology, Faculty of Biological Sciences, University of Concepción, Chile
| | - Wolfgang Schmidt-Heck
- Leibniz Institute for Natural Product Research and Infection Biology eV-Hans-Knöll Institute, Jena, Germany
| | - Karthick Natarajan
- University of Cologne, Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | | | - Dagmara Szkolnicka
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - Annika Asplund
- Takara Bio Europe AB (former Cellartis AB), Arvid Wallgrens Backe 20, 41346 Gothenburg, Sweden; Systems Biology Research Center, School of Bioscience, University of Skövde, Sweden
| | | | - Agata Widera
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - Regina Stöber
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - Gisela Campos
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - Seddik Hammad
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - Agapios Sachinidis
- University of Cologne, Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Umesh Chaudhari
- University of Cologne, Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Georg Damm
- Charité University Medicine Berlin, Department of General-, Visceral- and Transplantation Surgery, D13353 Berlin, Germany
| | - Thomas S Weiss
- Center for Liver Cell Research, Department of Pediatrics and Juvenile Medicine, University of Regensburg Hospital, Regensburg, Germany
| | - Andreas Nüssler
- Eberhard Karls University Tübingen, BG Trauma Center, Siegfried Weller Institut, D72076 Tübingen, Germany
| | - Jane Synnergren
- Systems Biology Research Center, School of Bioscience, University of Skövde, Sweden
| | - Karolina Edlund
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - Barbara Küppers-Munther
- Takara Bio Europe AB (former Cellartis AB), Arvid Wallgrens Backe 20, 41346 Gothenburg, Sweden
| | - David C Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom.
| | - Jan G Hengstler
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany.
| |
Collapse
|
152
|
Villarin BL, Cameron K, Szkolnicka D, Rashidi H, Bates N, Kimber SJ, Flint O, Forbes SJ, Iredale JP, Bradley M, Hay DC. Polymer Supported Directed Differentiation Reveals a Unique Gene Signature Predicting Stable Hepatocyte Performance. Adv Healthc Mater 2015; 4:1820-5. [PMID: 26109270 DOI: 10.1002/adhm.201500391] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Indexed: 01/14/2023]
Abstract
In theory, pluripotent stem cells can give rise to all somatic cell types found in the human body. The ability to generate renewable sources of human cells has enormous potential to improve human health and wealth. One major obstacle to the routine deployment of stem cell-derived cells is their instability in culture. To tackle this issue a synthetic polymer surface is used.
Collapse
Affiliation(s)
- Baltasar Lucendo Villarin
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Kate Cameron
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Dagmara Szkolnicka
- 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
| | - Nicola Bates
- Faculty of Life Sciences, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Susan J Kimber
- Faculty of Life Sciences, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Oliver Flint
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Stuart J Forbes
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - John P Iredale
- MRC Centre for Inflammation Research, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Mark Bradley
- School of Chemistry, University of Edinburgh, West Mains Road, Edinburgh, EH9 3FJ, UK
| | - David C Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| |
Collapse
|
153
|
Liu J, Brzeszczynska J, Samuel K, Black J, Palakkan A, Anderson RA, Gallagher R, Ross JA. Efficient episomal reprogramming of blood mononuclear cells and differentiation to hepatocytes with functional drug metabolism. Exp Cell Res 2015; 338:203-13. [PMID: 26256888 DOI: 10.1016/j.yexcr.2015.08.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/04/2015] [Accepted: 08/06/2015] [Indexed: 12/12/2022]
Abstract
The possibility of converting cells from blood mononuclear cells (MNC) to liver cells provides promising opportunities for the study of diseases and the assessment of new drugs. However, clinical applications have to meet GMP requirements and the methods for generating induced pluripotent cells (iPCs) have to avoid insertional mutagenesis, a possibility when using viral vehicles for the delivery of reprogramming factors. We have developed an efficient non-integration method for reprogramming fresh or frozen blood MNC, maintained in an optimised cytokine cocktail, to generate induced pluripotent cells. Using electroporation for the effective delivery of episomal transcription factors (Oct4, Sox2, Klf4, L-Myc, and Lin28) in a feeder-free system, without any requirement for small molecules, we achieved a reprogramming efficiency of up to 0.033% (65 colonies from 2×10(5) seeded MNC). Applying the same cytokine cocktail and reprogramming methods to cord blood or fetal liver-derived CD34(+) cells, we obtained 148 iPS colonies from 10(5) seeding cells (0.148%). The iPS cell lines we generated maintained typical characteristics of pluripotent cells and could be successfully differentiated into hepatocytes with drug metabolic function.
Collapse
Affiliation(s)
- Jing Liu
- Scottish National Blood Transfusion Service (SNBTS) Cell Therapy Group, Centre for Regenerative Medicine, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, United Kingdom; Tissue Injury and Repair Group, MRC Centre for Regenerative Medicine, University of Edinburgh, EH16 4SB United Kingdom; Peking University Institute of Hematology, Peking University People's Hospital, Beijing, 100044 China
| | - Joanna Brzeszczynska
- Tissue Injury and Repair Group, MRC Centre for Regenerative Medicine, University of Edinburgh, EH16 4SB United Kingdom
| | - Kay Samuel
- Scottish National Blood Transfusion Service (SNBTS) Cell Therapy Group, Centre for Regenerative Medicine, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, United Kingdom
| | - Jim Black
- Tissue Injury and Repair Group, MRC Centre for Regenerative Medicine, University of Edinburgh, EH16 4SB United Kingdom
| | - Anwar Palakkan
- Tissue Injury and Repair Group, MRC Centre for Regenerative Medicine, University of Edinburgh, EH16 4SB United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, United Kingdom
| | - Ronald Gallagher
- Scottish National Blood Transfusion Service (SNBTS) Cell Therapy Group, Centre for Regenerative Medicine, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, United Kingdom.
| | - James A Ross
- Tissue Injury and Repair Group, MRC Centre for Regenerative Medicine, University of Edinburgh, EH16 4SB United Kingdom.
| |
Collapse
|
154
|
Tasnim F, Phan D, Toh YC, Yu H. Cost-effective differentiation of hepatocyte-like cells from human pluripotent stem cells using small molecules. Biomaterials 2015; 70:115-25. [PMID: 26310107 DOI: 10.1016/j.biomaterials.2015.08.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 08/01/2015] [Indexed: 12/20/2022]
Abstract
Significant efforts have been invested into the differentiation of stem cells into functional hepatocyte-like cells that can be used for cell therapy, disease modeling and drug screening. Most of these efforts have been concentrated on the use of growth factors to recapitulate developmental signals under in vitro conditions. Using small molecules instead of growth factors would provide an attractive alternative since small molecules are cell-permeable and cheaper than growth factors. We have developed a protocol for the differentiation of human embryonic stem cells into hepatocyte-like cells using a predominantly small molecule-based approach (SM-Hep). This 3 step differentiation strategy involves the use of optimized concentrations of LY294002 and bromo-indirubin-3'-oxime (BIO) for the generation of definitive endoderm; sodium butyrate and dimethyl sulfoxide (DMSO) for the generation of hepatoblasts and SB431542 for differentiation into hepatocyte-like cells. Activin A is the only growth factor required in this protocol. Our results showed that SM-Hep were morphologically and functionally similar or better compared to the hepatocytes derived from the growth-factor induced differentiation (GF-Hep) in terms of expression of hepatic markers, urea and albumin production and cytochrome P450 (CYP1A2 and CYP3A4) activities. Cell viability assays following treatment with paradigm hepatotoxicants Acetaminophen, Chlorpromazine, Diclofenac, Digoxin, Quinidine and Troglitazone showed that their sensitivity to these drugs was similar to human primary hepatocytes (PHHs). Using SM-Hep would result in 67% and 81% cost reduction compared to GF-Hep and PHHs respectively. Therefore, SM-Hep can serve as a robust and cost effective replacement for PHHs for drug screening and development.
Collapse
Affiliation(s)
- Farah Tasnim
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Derek Phan
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Yi-Chin Toh
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Hanry Yu
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, MD9-03-03, 2 Medical Drive, Singapore 117597, Singapore; NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences, #05-01, 28 Medical Drive, Singapore 117576, Singapore; Mechanobiology Institute, T-Labs, #05-01, 5A Engineering Drive 1, Singapore 117411, Singapore; Singapore-MIT Alliance for Research and Technology, 3 Science Drive 2, S16-05-08, Singapore 117543, Singapore; Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA.
| |
Collapse
|
155
|
Lu WY, Bird TG, Boulter L, Tsuchiya A, Cole AM, Hay T, Guest RV, Wojtacha D, Man TY, Mackinnon A, Ridgway RA, Kendall T, Williams MJ, Jamieson T, Raven A, Hay DC, Iredale JP, Clarke AR, Sansom OJ, Forbes SJ. Hepatic progenitor cells of biliary origin with liver repopulation capacity. Nat Cell Biol 2015; 17:971-983. [PMID: 26192438 PMCID: PMC4612439 DOI: 10.1038/ncb3203] [Citation(s) in RCA: 365] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/09/2015] [Indexed: 12/13/2022]
Abstract
Hepatocytes and cholangiocytes self-renew following liver injury. Following severe injury hepatocytes are increasingly senescent, but whether hepatic progenitor cells (HPCs) then contribute to liver regeneration is unclear. Here, we describe a mouse model where the E3 ubiquitin ligase Mdm2 is inducibly deleted in more than 98% of hepatocytes, causing apoptosis, necrosis and senescence with nearly all hepatocytes expressing p21. This results in florid HPC activation, which is necessary for survival, followed by complete, functional liver reconstitution. HPCs isolated from genetically normal mice, using cell surface markers, were highly expandable and phenotypically stable in vitro. These HPCs were transplanted into adult mouse livers where hepatocyte Mdm2 was repeatedly deleted, creating a non-competitive repopulation assay. Transplanted HPCs contributed significantly to restoration of liver parenchyma, regenerating hepatocytes and biliary epithelia, highlighting their in vivo lineage potency. HPCs are therefore a potential future alternative to hepatocyte or liver transplantation for liver disease.
Collapse
Affiliation(s)
- Wei-Yu Lu
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - Thomas G Bird
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Edinburgh, EH4 2XU
| | - Atsunori Tsuchiya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, Niigata, Japan
| | - Alicia M Cole
- The CRUK Beatson Institute for Cancer Research, Switchback Road, Bearsden, Glasgow, G61 1BD
| | - Trevor Hay
- European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, CF24 4HQ
| | - Rachel V Guest
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - Davina Wojtacha
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - Tak Yung Man
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - Alison Mackinnon
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - Rachel A Ridgway
- The CRUK Beatson Institute for Cancer Research, Switchback Road, Bearsden, Glasgow, G61 1BD
| | - Timothy Kendall
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Edinburgh, EH4 2XU
| | - Michael J Williams
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - Thomas Jamieson
- The CRUK Beatson Institute for Cancer Research, Switchback Road, Bearsden, Glasgow, G61 1BD
| | - Alex Raven
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - David C Hay
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| | - John P Iredale
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Alan R Clarke
- European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, CF24 4HQ
| | - Owen J Sansom
- The CRUK Beatson Institute for Cancer Research, Switchback Road, Bearsden, Glasgow, G61 1BD
| | - Stuart J Forbes
- MRC Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh, EH16 4UU
| |
Collapse
|
156
|
Signaling Control of Differentiation of Embryonic Stem Cells toward Mesendoderm. J Mol Biol 2015; 428:1409-22. [PMID: 26119455 DOI: 10.1016/j.jmb.2015.06.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 06/12/2015] [Accepted: 06/17/2015] [Indexed: 01/29/2023]
Abstract
Mesendoderm (ME) refers to the primitive streak in mammalian embryos, which has the ability to further differentiate into mesoderm and endoderm. A better understanding on the regulatory networks of ME differentiation of embryonic stem (ES) cells would provide important insights on early embryo patterning and a possible guidance for ES applications in regenerative medicine. Studies on developmental biology and embryology have offered a great deal of knowledge about key signaling pathways involved in primitive streak formation. Recently, various chemically defined recipes have been formulated to induce differentiation of ES cells toward ME in vitro, which greatly facilitate the elucidation of the regulatory mechanisms of different signals involved in ME specification. Among the extrinsic signals, transforming growth factor-β/Activin signaling and Wnt signaling have been shown to be the most critical ones. On another side, intrinsic epigenetic regulation has been indicated to be important in ME determination. In this review, we summarize the current understanding on the extrinsic and intrinsic regulations of ES cells-to-ME differentiation and the crosstalk among them, aiming to get a general overview on ME specification and primitive streak formation.
Collapse
|
157
|
Moulin F, Flint O. In VitroModels for the Prediction of Drug-Induced Liver Injury in Lead Discovery. ACTA ACUST UNITED AC 2015. [DOI: 10.1002/9783527673643.ch07] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
158
|
Kim JH, Jang YJ, An SY, Son J, Lee J, Lee G, Park JY, Park HJ, Hwang DY, Kim JH, Han J. Enhanced Metabolizing Activity of Human ES Cell-Derived Hepatocytes Using a 3D Culture System with Repeated Exposures to Xenobiotics. Toxicol Sci 2015; 147:190-206. [DOI: 10.1093/toxsci/kfv121] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
159
|
Pettinato G, Wen X, Zhang N. Engineering Strategies for the Formation of Embryoid Bodies from Human Pluripotent Stem Cells. Stem Cells Dev 2015; 24:1595-609. [PMID: 25900308 DOI: 10.1089/scd.2014.0427] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are powerful tools for regenerative therapy and studying human developmental biology, attributing to their ability to differentiate into many functional cell types in the body. The main challenge in realizing hPSC potential is to guide their differentiation in a well-controlled manner. One way to control the cell differentiation process is to recapitulate during in vitro culture the key events in embryogenesis to obtain the three developmental germ layers from which all cell types arise. To achieve this goal, many techniques have been tested to obtain a cellular cluster, an embryoid body (EB), from both mouse and hPSCs. Generation of EBs that are homogeneous in size and shape would allow directed hPSC differentiation into desired cell types in a more synchronous manner and define the roles of cell-cell interaction and spatial organization in lineage specification in a setting similar to in vivo embryonic development. However, previous success in uniform EB formation from mouse PSCs cannot be extrapolated to hPSCs possibly due to the destabilization of adherens junctions on cell surfaces during the dissociation into single cells, making hPSCs extremely vulnerable to cell death. Recently, new advances have emerged to form uniform human embryoid bodies (hEBs) from dissociated single cells of hPSCs. In this review, the existing methods for hEB production from hPSCs and the results on the downstream differentiation of the hEBs are described with emphases on the efficiency, homogeneity, scalability, and reproducibility of the hEB formation process and the yield in terminal differentiation. New trends in hEB production and directed differentiation are discussed.
Collapse
Affiliation(s)
- Giuseppe Pettinato
- 1 Department of Biomedical Engineering, Virginia Commonwealth University , Richmond, Virginia.,2 Department of Chemical and Life Science Engineering, Virginia Commonwealth University , Richmond, Virginia
| | - Xuejun Wen
- 2 Department of Chemical and Life Science Engineering, Virginia Commonwealth University , Richmond, Virginia.,3 Shanghai East Hospital, The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Ning Zhang
- 1 Department of Biomedical Engineering, Virginia Commonwealth University , Richmond, Virginia
| |
Collapse
|
160
|
Giobbe GG, Michielin F, Luni C, Giulitti S, Martewicz S, Dupont S, Floreani A, Elvassore N. Functional differentiation of human pluripotent stem cells on a chip. Nat Methods 2015; 12:637-40. [DOI: 10.1038/nmeth.3411] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 03/10/2015] [Indexed: 12/20/2022]
|
161
|
Siller R, Greenhough S, Naumovska E, Sullivan GJ. Small-molecule-driven hepatocyte differentiation of human pluripotent stem cells. Stem Cell Reports 2015; 4:939-52. [PMID: 25937370 PMCID: PMC4437467 DOI: 10.1016/j.stemcr.2015.04.001] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 04/02/2015] [Accepted: 04/02/2015] [Indexed: 12/17/2022] Open
Abstract
The differentiation of pluripotent stem cells to hepatocytes is well established, yet current methods suffer from several drawbacks. These include a lack of definition and reproducibility, which in part stems from continued reliance on recombinant growth factors. This has remained a stumbling block for the translation of the technology into industry and the clinic for reasons associated with cost and quality. We have devised a growth-factor-free protocol that relies on small molecules to differentiate human pluripotent stem cells toward a hepatic phenotype. The procedure can efficiently direct both human embryonic stem cells and induced pluripotent stem cells to hepatocyte-like cells. The final population of cells demonstrates marker expression at the transcriptional and protein levels, as well as key hepatic functions such as serum protein production, glycogen storage, and cytochrome P450 activity. Development of small-molecule-driven hepatocyte differentiation procedure for hPSCs Small-molecule-derived hepatocytes demonstrate key hepatic functions Significantly reduces the cost of hepatocyte differentiation Procedure is applicable to multiple human pluripotent stem cell lines
Collapse
Affiliation(s)
- Richard Siller
- Department of Biochemistry, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, PO Box 1112 Blindern, 0317 Oslo, Norway
| | - Sebastian Greenhough
- Department of Biochemistry, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, PO Box 1112 Blindern, 0317 Oslo, Norway
| | - Elena Naumovska
- Department of Biochemistry, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, PO Box 1112 Blindern, 0317 Oslo, Norway
| | - Gareth J Sullivan
- Department of Biochemistry, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, PO Box 1112 Blindern, 0317 Oslo, Norway; Norwegian Center for Stem Cell Research, PO Box 1112 Blindern, 0317 Oslo, Norway; Institute of Immunology, Oslo University Hospital-Rikshospitalet, PO Box 4950 Nydalen, Oslo 0424, Norway.
| |
Collapse
|
162
|
Forbes SJ, Gupta S, Dhawan A. Cell therapy for liver disease: From liver transplantation to cell factory. J Hepatol 2015; 62:S157-69. [PMID: 25920085 DOI: 10.1016/j.jhep.2015.02.040] [Citation(s) in RCA: 218] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 02/20/2015] [Accepted: 02/27/2015] [Indexed: 02/08/2023]
Abstract
Work over several decades has laid solid foundations for the advancement of liver cell therapy. To date liver cell therapy in people has taken the form of hepatocyte transplantation for metabolic disorders with a hepatic basis, and for acute or chronic liver failure. Although clinical trials using various types of autologous cells have been implemented to promote liver regeneration or reduce liver fibrosis, clear evidence of therapeutic benefits have so far been lacking. Cell types that have shown efficacy in preclinical models include hepatocytes, liver sinusoidal endothelial cells, mesenchymal stem cells, endothelial progenitor cells, and macrophages. However, positive results in animal models have not always translated through to successful clinical therapies and more realistic preclinical models need to be developed. Studies defining the optimal repopulation by transplanted cells, including routes of cell transplantation, superior engraftment and proliferation of transplanted cells, as well as optimal immunosuppression regimens are required. Tissue engineering approaches to transplant cells in extrahepatic locations have also been proposed. The derivation of hepatocytes from pluripotent or reprogrammed cells raises hope that donor organ and cell shortages could be overcome in the future. Critical hurdles to be overcome include the production of hepatocytes from pluripotent cells with equal functional capacity to primary hepatocytes and long-term phenotypic stability in vivo.
Collapse
Affiliation(s)
- Stuart J Forbes
- MRC Centre for Regenerative Medicine, Scottish Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh EH16 4UU, United Kingdom.
| | - Sanjeev Gupta
- Departments of Medicine and Pathology, Albert Einstein College of Medicine, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Ullmann Building, Room 625, Bronx, NY 10461, United States
| | - Anil Dhawan
- Paediatric Liver GI and Nutrition Center and NIHR/Wellcome Cell Therapy Unit, King's College Hospital at King's College, London SE59RS, United Kingdom
| |
Collapse
|
163
|
Suter-Dick L, Alves PM, Blaauboer BJ, Bremm KD, Brito C, Coecke S, Flick B, Fowler P, Hescheler J, Ingelman-Sundberg M, Jennings P, Kelm JM, Manou I, Mistry P, Moretto A, Roth A, Stedman D, van de Water B, Beilmann M. Stem cell-derived systems in toxicology assessment. Stem Cells Dev 2015; 24:1284-96. [PMID: 25675366 DOI: 10.1089/scd.2014.0540] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Industrial sectors perform toxicological assessments of their potential products to ensure human safety and to fulfill regulatory requirements. These assessments often involve animal testing, but ethical, cost, and time concerns, together with a ban on it in specific sectors, make appropriate in vitro systems indispensable in toxicology. In this study, we summarize the outcome of an EPAA (European Partnership of Alternatives to Animal Testing)-organized workshop on the use of stem cell-derived (SCD) systems in toxicology, with a focus on industrial applications. SCD systems, in particular, induced pluripotent stem cell-derived, provide physiological cell culture systems of easy access and amenable to a variety of assays. They also present the opportunity to apply the vast repository of existing nonclinical data for the understanding of in vitro to in vivo translation. SCD systems from several toxicologically relevant tissues exist; they generally recapitulate many aspects of physiology and respond to toxicological and pharmacological interventions. However, focused research is necessary to accelerate implementation of SCD systems in an industrial setting and subsequent use of such systems by regulatory authorities. Research is required into the phenotypic characterization of the systems, since methods and protocols for generating terminally differentiated SCD cells are still lacking. Organotypical 3D culture systems in bioreactors and microscale tissue engineering technologies should be fostered, as they promote and maintain differentiation and support coculture systems. They need further development and validation for their successful implementation in toxicity testing in industry. Analytical measures also need to be implemented to enable compound exposure and metabolism measurements for in vitro to in vivo extrapolation. The future of SCD toxicological tests will combine advanced cell culture technologies and biokinetic measurements to support regulatory and research applications. However, scientific and technical hurdles must be overcome before SCD in vitro methods undergo appropriate validation and become accepted in the regulatory arena.
Collapse
Affiliation(s)
- Laura Suter-Dick
- 1University of Applied Sciences Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland
| | - Paula M Alves
- 2iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,3Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Bas J Blaauboer
- 4Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| | - Klaus-Dieter Bremm
- 5Bayer Pharma AG, Global Drug Discovery-Global Early Development, Wuppertal, Germany
| | - Catarina Brito
- 2iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,3Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Sandra Coecke
- 6European Commission Joint Research Centre, Institute for Health and Consumer Protection, EURL ECVAM, Ispra, Italy
| | - Burkhard Flick
- 7BASF SE, Experimental Toxicology and Ecology, Ludwigshafen, Germany
| | | | - Jürgen Hescheler
- 9Institut for Neurophysiology, University of Cologne, Cologne, Germany
| | | | - Paul Jennings
- 11Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Innsbruck, Austria
| | | | - Irene Manou
- 13European Partnership for Alternative Approaches to Animal Testing (EPAA), B-Brussels, Belgium
| | - Pratibha Mistry
- 14Syngenta Ltd., Product Safety, Jealott's Hill International Research Station, Berkshire, United Kingdom
| | - Angelo Moretto
- 15Dipartimento di Scienze Biochimiche e Cliniche, Università degli Studi di Milano, Milano, Italy.,16Centro Internazionale per gli Antiparassitari e la Prevenzione Sanitaria, Luigi Sacco Hospital, Milano, Italy
| | - Adrian Roth
- 17F. Hoffmann-La Roche Ltd., Innovation Center Basel, Pharmaceutical Sciences, Basel, Switzerland
| | - Donald Stedman
- 18Pfizer Worldwide Research and Development, Cambridge, Massachusetts
| | - Bob van de Water
- 19Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | | |
Collapse
|
164
|
Pezzolla D, López-Beas J, Lachaud CC, Domínguez-Rodríguez A, Smani T, Hmadcha A, Soria B. Resveratrol ameliorates the maturation process of β-cell-like cells obtained from an optimized differentiation protocol of human embryonic stem cells. PLoS One 2015; 10:e0119904. [PMID: 25774684 PMCID: PMC4361612 DOI: 10.1371/journal.pone.0119904] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/03/2015] [Indexed: 12/11/2022] Open
Abstract
Human embryonic stem cells (hESCs) retain the extraordinary capacity to differentiate into different cell types of an adult organism, including pancreatic β-cells. For this particular lineage, although a lot of effort has been made in the last ten years to achieve an efficient and reproducible differentiation protocol, it was not until recently that this aim was roughly accomplished. Besides, several studies evidenced the impact of resveratrol (RSV) on insulin secretion, even though the mechanism by which this polyphenol potentiates glucose-stimulated insulin secretion (GSIS) is still not clear. The aim of this study was to optimize an efficient differentiation protocol that mimics in vivo pancreatic organogenesis and to investigate whether RSV may improve the final maturation step to obtain functional insulin-secreting cells. Our results indicate that treatment of hESCs (HS-181) with activin-A induced definitive endoderm differentiation as detected by the expression of SOX17 and FOXA2. Addition of retinoic acid (RA), Noggin and Cyclopamine promoted pancreatic differentiation as indicated by the expression of the early pancreatic progenitor markers ISL1, NGN3 and PDX1. Moreover, during maturation in suspension culture, differentiating cells assembled in islet-like clusters, which expressed specific endocrine markers such as PDX1, SST, GCG and INS. Similar results were confirmed with the human induced Pluripotent Stem Cell (hiPSC) line MSUH-001. Finally, differentiation protocols incorporating RSV treatment yielded numerous insulin-positive cells, induced significantly higher PDX1 expression and were able to transiently normalize glycaemia when transplanted in streptozotocin (STZ) induced diabetic mice thus promoting its survival. In conclusion, our strategy allows the efficient differentiation of hESCs into pancreatic endoderm capable of generating β-cell-like cells and demonstrates that RSV improves the maturation process.
Collapse
Affiliation(s)
- Daniela Pezzolla
- Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)—Fundación Progreso y Salud (FPS), Sevilla, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Barcelona, Spain
| | - Javier López-Beas
- Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)—Fundación Progreso y Salud (FPS), Sevilla, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Barcelona, Spain
| | - Christian C. Lachaud
- Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)—Fundación Progreso y Salud (FPS), Sevilla, Spain
| | | | - Tarik Smani
- Cardiovascular Pathophysiology, Institute of Biomedicine of Seville (IBIS), Sevilla, Spain
| | - Abdelkrim Hmadcha
- Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)—Fundación Progreso y Salud (FPS), Sevilla, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Barcelona, Spain
- * E-mail:
| | - Bernat Soria
- Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)—Fundación Progreso y Salud (FPS), Sevilla, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Barcelona, Spain
| |
Collapse
|
165
|
Tsankov AM, Gu H, Akopian V, Ziller MJ, Donaghey J, Amit I, Gnirke A, Meissner A. Transcription factor binding dynamics during human ES cell differentiation. Nature 2015; 518:344-9. [PMID: 25693565 PMCID: PMC4499331 DOI: 10.1038/nature14233] [Citation(s) in RCA: 268] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/14/2015] [Indexed: 12/24/2022]
Abstract
Pluripotent stem cells provide a powerful system to dissect the underlying molecular dynamics that regulate cell fate changes during mammalian development. Here we report the integrative analysis of genome-wide binding data for 38 transcription factors with extensive epigenome and transcriptional data across the differentiation of human embryonic stem cells to the three germ layers. We describe core regulatory dynamics and show the lineage-specific behaviour of selected factors. In addition to the orchestrated remodelling of the chromatin landscape, we find that the binding of several transcription factors is strongly associated with specific loss of DNA methylation in one germ layer, and in many cases a reciprocal gain in the other layers. Taken together, our work shows context-dependent rewiring of transcription factor binding, downstream signalling effectors, and the epigenome during human embryonic stem cell differentiation.
Collapse
Affiliation(s)
- Alexander M Tsankov
- 1] Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA [2] Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA [3] Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Hongcang Gu
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Veronika Akopian
- 1] Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA [2] Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Michael J Ziller
- 1] Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA [2] Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA [3] Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Julie Donaghey
- 1] Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA [2] Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA [3] Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Ido Amit
- 1] Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA [2] Department of Immunology, Weizmann Institute, Rehovot, 76100 Israel
| | - Andreas Gnirke
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Alexander Meissner
- 1] Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA [2] Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA [3] Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| |
Collapse
|
166
|
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: 253] [Impact Index Per Article: 25.3] [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
|
167
|
Ikonomou L, Kotton DN. Derivation of Endodermal Progenitors From Pluripotent Stem Cells. J Cell Physiol 2015; 230:246-58. [PMID: 25160562 PMCID: PMC4344429 DOI: 10.1002/jcp.24771] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 08/22/2014] [Indexed: 01/18/2023]
Abstract
Stem and progenitor cells play important roles in organogenesis during development and in tissue homeostasis and response to injury postnatally. As the regenerative capacity of many human tissues is limited, cell replacement therapies hold great promise for human disease management. Pluripotent stem cells such as embryonic stem (ES) cells and induced pluripotent stem (iPS) cells are prime candidates for the derivation of unlimited quantities of clinically relevant cell types through development of directed differentiation protocols, that is, the recapitulation of developmental milestones in in vitro cell culture. Tissue-specific progenitors, including progenitors of endodermal origin, are important intermediates in such protocols since they give rise to all mature parenchymal cells. In this review, we focus on the in vivo biology of embryonic endodermal progenitors in terms of key transcription factors and signaling pathways. We critically review the emerging literature aiming to apply this basic knowledge to achieve the efficient and reproducible in vitro derivation of endodermal progenitors such as pancreas, liver and lung precursor cells.
Collapse
Affiliation(s)
- Laertis Ikonomou
- Center for Regenerative Medicine, Boston University and Boston
Medical Center, Boston, MA, USA
- Boston University Pulmonary Center, Boston University School of
Medicine, Boston, MA, USA
| | - Darrell N. Kotton
- Center for Regenerative Medicine, Boston University and Boston
Medical Center, Boston, MA, USA
- Boston University Pulmonary Center, Boston University School of
Medicine, Boston, MA, USA
| |
Collapse
|
168
|
Schotanus BA, Kruitwagen HS, van den Ingh TSGAM, van Wolferen ME, Rothuizen J, Penning LC, Spee B. Enhanced Wnt/β-catenin and Notch signalling in the activated canine hepatic progenitor cell niche. BMC Vet Res 2014; 10:309. [PMID: 25551829 PMCID: PMC4302101 DOI: 10.1186/s12917-014-0309-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/16/2014] [Indexed: 02/06/2023] Open
Abstract
Background The liver has a large regenerative capacity. Hepatocytes can replicate and regenerate a diseased liver. However, as is the case in severe liver diseases, this replication may become insufficient or exhausted and hepatic progenitor cells (HPCs) can be activated in an attempt to restore liver function. Due to their bi-potent differentiation capacity, these HPCs have great potential for regenerative approaches yet over-activation does pose potential health risks. Therefore the mechanisms leading to activation must be elucidated prior to safe implementation in the veterinary clinic. Wnt/β-catenin and Notch signalling have been implicated in the activation of HPCs in mouse models and in humans. Here we assessed the involvement in canine HPC activation. Gene-expression profiles were derived from laser microdissected HPC niches from lobular dissecting hepatitis (LDH) and normal liver tissue, with a focus on Wnt/β-catenin and Notch signalling. Immunohistochemical and immunofluorescent studies were combined to assess the role of the pathways in HPCs during LDH. Results Gene-expression confirmed higher expression of Wnt/β-catenin and Notch pathway components and target genes in activated HPC niches in diseased liver compared to quiescent HPC niches from normal liver. Immunofluorescence confirmed the activation of these pathways in the HPCs during disease. Immunohistochemistry showed proliferating HPCs during LDH, and double immunofluorescence showed downregulation of Wnt/β-catenin and Notch in differentiating HPCs. Vimentin, a mesenchymal marker, was expressed on a subset of undifferentiated HPCs. Conclusions Together these studies clearly revealed that both Wnt/β-catenin and Notch signalling pathways are enhanced in undifferentiated, proliferating and potentially migrating HPCs during severe progressive canine liver disease (LDH).
Collapse
Affiliation(s)
- Baukje A Schotanus
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Hedwig S Kruitwagen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | | | - Monique E van Wolferen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Jan Rothuizen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Louis C Penning
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
169
|
Goldman O, Han S, Sourisseau M, Sourrisseau M, Dziedzic N, Hamou W, Corneo B, D'Souza S, Sato T, Kotton DN, Bissig KD, Kalir T, Jacobs A, Evans T, Evans MJ, Gouon-Evans V. KDR identifies a conserved human and murine hepatic progenitor and instructs early liver development. Cell Stem Cell 2014; 12:748-60. [PMID: 23746980 DOI: 10.1016/j.stem.2013.04.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 08/10/2012] [Accepted: 04/29/2013] [Indexed: 01/22/2023]
Abstract
Understanding the fetal hepatic niche is essential for optimizing the generation of functional hepatocyte-like cells (hepatic cells) from human embryonic stem cells (hESCs). Here, we show that KDR (VEGFR2/FLK-1), previously assumed to be mostly restricted to mesodermal lineages, marks a hESC-derived hepatic progenitor. hESC-derived endoderm cells do not express KDR but, when cultured in media supporting hepatic differentiation, generate KDR+ hepatic progenitors and KDR- hepatic cells. KDR+ progenitors require active KDR signaling both to instruct their own differentiation into hepatic cells and to non-cell-autonomously support the functional maturation of cocultured KDR- hepatic cells. Analysis of human fetal livers suggests that similar progenitors are present in human livers. Lineage tracing in mice provides in vivo evidence of a KDR+ hepatic progenitor for fetal hepatoblasts, adult hepatocytes, and adult cholangiocytes. Altogether, our findings reveal that KDR is a conserved marker for endoderm-derived hepatic progenitors and a functional receptor instructing early liver development.
Collapse
Affiliation(s)
- Orit Goldman
- Department of Developmental and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Yao R, Wang J, Li X, Jung Jung D, Qi H, Kee KK, Du Y. Hepatic differentiation of human embryonic stem cells as microscaled multilayered colonies leading to enhanced homogeneity and maturation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:4311-4323. [PMID: 25059765 DOI: 10.1002/smll.201401040] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 05/28/2014] [Indexed: 06/03/2023]
Abstract
Directed differentiation of human embryonic stem cells (hESCs) towards hepatocyte-like cells on planar tissue culture plates has been extensively investigated with great promise to provide alternative cell sources for drug metabolism/toxicity testing. Recently, hepatic differentiation of hESCs in 3D configuration with better mimicry of embryonic liver development represents incremental efforts to improve the differentiation efficiency and cellular maturation. However, most of the present 3D differentiation configurations involved interruptive operations during the multi-staged differentiation process, which might impose unwanted influence on cellular differentiation. Most of the current researches resulted in generation of hepatocytes with high expression of AFP, which is minimally expressed in primary hepatocytes. Here, off-the-shelf micro-stencil arrays are developed to generate adherent multilayered colonies composed of hESCs-derived cells. Uninterrupted cellular differentiation and proliferation is achieved to recapitulate the continuous and multi-stage liver development. Compared with conventional 2D format, the micro-scaled multilayered colonies with uniform and defined sizes constrained within the microwells are composed of more homogenous and mature hepatocyte-like cells with significantly lowered AFP expression and elevated hepatic functions. The multilayered colonies as novel 3D configuration for hepatic differentiation of hESCs represent a significant step toward efficient generation of functional hepatocytes for regenerative medicine and drug discovery.
Collapse
Affiliation(s)
- Rui Yao
- Biofabrication Center, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China; Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | | | | | | | | | | | | |
Collapse
|
171
|
Semeraro R, Cardinale V, Carpino G, Gentile R, Napoli C, Venere R, Gatto M, Brunelli R, Gaudio E, Alvaro D. The fetal liver as cell source for the regenerative medicine of liver and pancreas. ANNALS OF TRANSLATIONAL MEDICINE 2014; 1:13. [PMID: 25332958 DOI: 10.3978/j.issn.2305-5839.2012.10.02] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 10/15/2012] [Indexed: 12/11/2022]
Abstract
Patients affected by liver diseases and diabetes mellitus are in need for sources of new cells to enable a better transition into clinic programs of cell therapy and regenerative medicine. In this setting, fetal liver is becoming the most promising and available source of cells. Fetal liver displays unique characteristics given the possibility to isolate cell populations with a wide spectrum of endodermal differentiation and, the co-existence of endodermal and mesenchymal-derived cells. Thus, the fetal liver is a unique and highly available cell source contemporarily candidate for the regenerative medicine of both liver and pancreas. The purpose of this review is to revise the recent literature on the different stem cells populations isolable from fetal liver and candidate to cell therapy of liver diseases and diabetes and to discuss advantages and limitation with respect to other cell sources.
Collapse
Affiliation(s)
- Rossella Semeraro
- 1 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, 2 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, 3 Department of Obstetrics and Gynecology, Sapienza University of Rome, Rome, Italy ; 4 Department of Health Sciences, University of Rome "Foro Italico", Rome, Italy ; 5 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy
| | - Vincenzo Cardinale
- 1 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, 2 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, 3 Department of Obstetrics and Gynecology, Sapienza University of Rome, Rome, Italy ; 4 Department of Health Sciences, University of Rome "Foro Italico", Rome, Italy ; 5 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy
| | - Guido Carpino
- 1 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, 2 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, 3 Department of Obstetrics and Gynecology, Sapienza University of Rome, Rome, Italy ; 4 Department of Health Sciences, University of Rome "Foro Italico", Rome, Italy ; 5 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy
| | - Raffaele Gentile
- 1 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, 2 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, 3 Department of Obstetrics and Gynecology, Sapienza University of Rome, Rome, Italy ; 4 Department of Health Sciences, University of Rome "Foro Italico", Rome, Italy ; 5 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy
| | - Cristina Napoli
- 1 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, 2 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, 3 Department of Obstetrics and Gynecology, Sapienza University of Rome, Rome, Italy ; 4 Department of Health Sciences, University of Rome "Foro Italico", Rome, Italy ; 5 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy
| | - Rosanna Venere
- 1 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, 2 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, 3 Department of Obstetrics and Gynecology, Sapienza University of Rome, Rome, Italy ; 4 Department of Health Sciences, University of Rome "Foro Italico", Rome, Italy ; 5 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy
| | - Manuela Gatto
- 1 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, 2 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, 3 Department of Obstetrics and Gynecology, Sapienza University of Rome, Rome, Italy ; 4 Department of Health Sciences, University of Rome "Foro Italico", Rome, Italy ; 5 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy
| | - Roberto Brunelli
- 1 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, 2 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, 3 Department of Obstetrics and Gynecology, Sapienza University of Rome, Rome, Italy ; 4 Department of Health Sciences, University of Rome "Foro Italico", Rome, Italy ; 5 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy
| | - Eugenio Gaudio
- 1 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, 2 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, 3 Department of Obstetrics and Gynecology, Sapienza University of Rome, Rome, Italy ; 4 Department of Health Sciences, University of Rome "Foro Italico", Rome, Italy ; 5 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy
| | - Domenico Alvaro
- 1 Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, 2 Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, 3 Department of Obstetrics and Gynecology, Sapienza University of Rome, Rome, Italy ; 4 Department of Health Sciences, University of Rome "Foro Italico", Rome, Italy ; 5 Eleonora Lorillard Spencer-Cenci Foundation, Rome, Italy
| |
Collapse
|
172
|
Carpentier A, Tesfaye A, Chu V, Nimgaonkar I, Zhang F, Lee SB, Thorgeirsson SS, Feinstone SM, Liang TJ. Engrafted human stem cell-derived hepatocytes establish an infectious HCV murine model. J Clin Invest 2014; 124:4953-64. [PMID: 25295540 DOI: 10.1172/jci75456] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 09/04/2014] [Indexed: 12/17/2022] Open
Abstract
The demonstrated ability to differentiate both human embryonic stem cells (hESCs) and patient-derived induced pluripotent stem cells (hiPSCs) into hepatocyte-like cells (HLCs) holds great promise for both regenerative medicine and liver disease research. Here, we determined that, despite an immature phenotype, differentiated HLCs are permissive to hepatitis C virus (HCV) infection and mount an interferon response to HCV infection in vitro. HLCs differentiated from hESCs and hiPSCs could be engrafted in the liver parenchyma of immune-deficient transgenic mice carrying the urokinase-type plasminogen activator gene driven by the major urinary protein promoter. The HLCs were maintained for more than 3 months in the livers of chimeric mice, in which they underwent further maturation and proliferation. These engrafted and expanded human HLCs were permissive to in vivo infection with HCV-positive sera and supported long-term infection of multiple HCV genotypes. Our study demonstrates efficient engraftment and in vivo HCV infection of human stem cell-derived hepatocytes and provides a model to study chronic HCV infection in patient-derived hepatocytes, action of antiviral therapies, and the biology of HCV infection.
Collapse
|
173
|
Lucendo-Villarin B, Cameron K, Szkolnicka D, Travers P, Khan F, Walton JG, Iredale J, Bradley M, Hay DC. Stabilizing hepatocellular phenotype using optimized synthetic surfaces. J Vis Exp 2014:51723. [PMID: 25285607 DOI: 10.3791/51723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Currently, one of the major limitations in cell biology is maintaining differentiated cell phenotype. Biological matrices are commonly used for culturing and maintaining primary and pluripotent stem cell derived hepatocytes. While biological matrices are useful, they permit short term culture of hepatocytes, limiting their widespread application. We have attempted to overcome the limitations using a synthetic polymer coating. Polymers represent one of the broadest classes of biomaterials and possess a wide range of mechanical, physical and chemical properties, which can be fine-tuned for purpose. Importantly, such materials can be scaled to quality assured standards and display batch-to-batch consistency. This is essential if cells are to be expanded for high through-put screening in the pharmaceutical testing industry or for cellular based therapy. Polyurethanes (PUs) are one group of materials that have shown promise in cell culture. Our recent progress in optimizing a polyurethane coated surface, for long-term culture of human hepatocytes displaying stable phenotype, is presented and discussed.
Collapse
Affiliation(s)
| | - Kate Cameron
- MRC Centre for Regenerative Medicine, University of Edinburgh
| | | | - Paul Travers
- MRC Centre for Regenerative Medicine, University of Edinburgh
| | | | | | - John Iredale
- MRC Centre for Inflammation Research, University of Edinburgh
| | | | - David C Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh;
| |
Collapse
|
174
|
Serio RN. Wnt of the Two Horizons: Putting Stem Cell Self-Renewal and Cell Fate Determination into Context. Stem Cells Dev 2014; 23:1975-90. [DOI: 10.1089/scd.2014.0055] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Ryan N. Serio
- Graduate School of Pharmacology, Weill Cornell Medical College, New York, New York
| |
Collapse
|
175
|
Szkolnicka D, Farnworth SL, Lucendo‐Villarin B, Hay DC. Deriving Functional Hepatocytes from Pluripotent Stem Cells. ACTA ACUST UNITED AC 2014; 30:1G.5.1-12. [DOI: 10.1002/9780470151808.sc01g05s30] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Dagmara Szkolnicka
- MRC Centre for Regenerative Medicine, University of Edinburgh Edinburgh United Kingdom
| | - Sarah L. Farnworth
- MRC Centre for Regenerative Medicine, University of Edinburgh Edinburgh United Kingdom
| | | | - David C. Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh Edinburgh United Kingdom
| |
Collapse
|
176
|
Zhou X, Sun P, Lucendo-Villarin B, Angus A, Szkolnicka D, Cameron K, Farnworth S, Patel A, Hay D. Modulating innate immunity improves hepatitis C virus infection and replication in stem cell-derived hepatocytes. Stem Cell Reports 2014; 3:204-14. [PMID: 25068132 PMCID: PMC4110790 DOI: 10.1016/j.stemcr.2014.04.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/28/2014] [Accepted: 04/29/2014] [Indexed: 02/05/2023] Open
Abstract
In this study, human embryonic stem cell-derived hepatocytes (hESC-Heps) were investigated for their ability to support hepatitis C virus (HCV) infection and replication. hESC-Heps were capable of supporting the full viral life cycle, including the release of infectious virions. Although supportive, hESC-Hep viral infection levels were not as great as those observed in Huh7 cells. We reasoned that innate immune responses in hESC-Heps may lead to the low level of infection and replication. Upon further investigation, we identified a strong type III interferon response in hESC-Heps that was triggered by HCV. Interestingly, specific inhibition of the JAK/STAT signaling pathway led to an increase in HCV infection and replication in hESC-Heps. Of note, the interferon response was not evident in Huh7 cells. In summary, we have established a robust cell-based system that allows the in-depth study of virus-host interactions in vitro.
Collapse
Affiliation(s)
- Xiaoling Zhou
- Shantou University Medical College, Shantou 515041, People’s Republic of China
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G11 5JR, UK
| | - Pingnan Sun
- Shantou University Medical College, Shantou 515041, People’s Republic of China
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G11 5JR, UK
| | | | - Allan G.N. Angus
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G11 5JR, UK
| | - Dagmara Szkolnicka
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Kate Cameron
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Sarah L. Farnworth
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Arvind H. Patel
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G11 5JR, UK
- Corresponding author
| | - David C. Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
- Corresponding author
| |
Collapse
|
177
|
Abstract
Stem cells constitute a population of "primitive cells" with the ability to divide indefinitely and give rise to specialized cells under special conditions. Because of these two characteristics they have received particular attention in recent decades. These cells are the primarily responsible factors for the regeneration of tissues and organs and for the healing of lesions, a feature that makes them a central key in the development of cell-based medicine, called Regenerative Medicine. The idea of wound and organ repair and body regeneration is as old as the mankind, reflecting the human desire for inhibiting aging and immortality and it is first described in the ancient Greek myth of Prometheus. It is of interest that the myth refers to liver, an organ with remarkable regenerative ability after loss of mass and function caused by liver injury or surgical resection. Over the last decade there has been an important progress in understanding liver physiology and the mechanisms underlying hepatic development and regeneration. As liver transplantation, despite its difficulties, remains the only effective therapy for advanced liver disease so far, scientific interest has nowadays been orientated towards Regenerative Medicine and the use of stem cells to repair damaged liver. This review is focused on the available literature concerning the role of stem cells in liver regeneration. It summarizes the results of studies concerning endogenous liver regeneration and stem cell experimental protocols. Moreover, this review discusses the clinical studies that have been conducted in humans so far.
Collapse
|
178
|
Comparative analysis reveals similarities between cultured submandibular salivary gland cells and liver progenitor cells. SPRINGERPLUS 2014; 3:183. [PMID: 24790827 PMCID: PMC4000360 DOI: 10.1186/2193-1801-3-183] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 04/03/2014] [Indexed: 01/20/2023]
Abstract
Mouse submandibular salivary gland cells and liver progenitor cells from long-term in vitro cultures with a high proliferation potential were side-by-side compared by methods of immunocytochemistry, quantitative real-time PCR, flow cytometry, and transcriptome analysis. The two cell types were found to be similar in expressing cell markers such as EpCAM, CD29, c-Kit, Sca-1, and c-Met. In addition, both cell types expressed cytokeratins 8, 18, and 19, alpha-fetoprotein, and (weakly) albumin. Unlike the liver cells, however, the salivary gland cells in culture showed high-level expression of cytokeratin 14 and CD49f, which was indicative of their origin from salivary gland ducts. Quantitative real-time PCR and deep-sequencing transcriptome analysis revealed similarities in the expression pattern of transcription factors between the two cell types. In this respect, however, the cultured salivary gland cells proved to be closer to exocrine cells of the pancreas than to the liver progenitor cells. Thus, ductal cells of postnatal submandibular salivary glands in culture show phenotypic convergence with progenitor cells of endodermal origin, suggesting that these glands may serve as a potential cell source for cellular therapy of hepatic and pancreatic disorders. The results of this study provide a deeper insight into the molecular features of salivary gland cells and may help optimize procedures for stimulating their differentiation in a specified direction.
Collapse
|
179
|
SOX2 Is Regulated Differently from NANOG and OCT4 in Human Embryonic Stem Cells during Early Differentiation Initiated with Sodium Butyrate. Stem Cells Int 2014; 2014:298163. [PMID: 24707296 PMCID: PMC3951062 DOI: 10.1155/2014/298163] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 01/07/2014] [Indexed: 01/04/2023] Open
Abstract
Transcription factors NANOG, OCT4, and SOX2 regulate self-renewal and pluripotency in human embryonic stem (hES) cells; however, their expression profiles during early differentiation of hES cells are unclear. In this study, we used multiparameter flow cytometric assay to detect all three transcription factors (NANOG, OCT4, and SOX2) simultaneously at single cell level and monitored the changes in their expression during early differentiation towards endodermal lineage (induced by sodium butyrate). We observed at least four distinct populations of hES cells, characterized by specific expression patterns of NANOG, OCT4, and SOX2 and differentiation markers. Our results show that a single cell can express both differentiation and pluripotency markers at the same time, indicating a gradual mode of developmental transition in these cells. Notably, distinct regulation of SOX2 during early differentiation events was detected, highlighting the potential importance of this transcription factor for self-renewal of hES cells during differentiation.
Collapse
|
180
|
Schwartz RE, Fleming HE, Khetani SR, Bhatia SN. Pluripotent stem cell-derived hepatocyte-like cells. Biotechnol Adv 2014; 32:504-13. [PMID: 24440487 DOI: 10.1016/j.biotechadv.2014.01.003] [Citation(s) in RCA: 205] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Revised: 01/03/2014] [Accepted: 01/06/2014] [Indexed: 12/13/2022]
Abstract
Liver disease is an important clinical problem, impacting over 30 million Americans and over 600 million people worldwide. It is the 12th leading cause of death in the United States and the 16th worldwide. Due to a paucity of donor organs, several thousand Americans die yearly while waiting for liver transplantation. Unfortunately, alternative tissue sources such as fetal hepatocytes and hepatic cell lines are unreliable, difficult to reproduce, and do not fully recapitulate hepatocyte phenotype and functions. As a consequence, alternative cell sources that do not have these limitations have been sought. Human embryonic stem (hES) cell- and induced pluripotent stem (iPS) cell-derived hepatocyte-like cells may enable cell based therapeutics, the study of the mechanisms of human disease and human development, and provide a platform for screening the efficacy and toxicity of pharmaceuticals. iPS cells can be differentiated in a step-wise fashion with high efficiency and reproducibility into hepatocyte-like cells that exhibit morphologic and phenotypic characteristics of hepatocytes. In addition, iPS-derived hepatocyte-like cells (iHLCs) possess some functional hepatic activity as they secrete urea, alpha-1-antitrypsin, and albumin. However, the combined phenotypic and functional traits exhibited by iHLCs resemble a relatively immature hepatic phenotype that more closely resembles that of fetal hepatocytes rather than adult hepatocytes. Specifically, iHLCs express fetal markers such as alpha-fetoprotein and lack key mature hepatocyte functions, as reflected by drastically reduced activity (~0.1%) of important detoxification enzymes (i.e. CYP2A6, CYP3A4). These key differences between iHLCs and primary adult human hepatocytes have limited the use of stem cells as a renewable source of functional adult hepatocytes for in vitro and in vivo applications. Unfortunately, the developmental pathways that control hepatocyte maturation from a fetal into an adult hepatocyte are poorly understood, which has hampered the field in its efforts to induce further maturation of iPS-derived hepatic lineage cells. This review analyzes recent developments in the derivation of hepatocyte-like cells, and proposes important points to consider and assays to perform during their characterization. In the future, we envision that iHLCs will be used as in vitro models of human disease, and in the longer term, provide an alternative cell source for drug testing and clinical therapy.
Collapse
Affiliation(s)
- R E Schwartz
- Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Medicine, Brigham and Women's Hospital, USA
| | - H E Fleming
- Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - S R Khetani
- Mechanical and Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - S N Bhatia
- Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Medicine, Brigham and Women's Hospital, USA.
| |
Collapse
|
181
|
Kuai XL, Shao N, Lu H, Xiao SD, Zheng Q. Differentiation of nonhuman primate embryonic stem cells into hepatocyte-like cells. J Dig Dis 2014; 15:27-34. [PMID: 24112234 DOI: 10.1111/1751-2980.12103] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate whether cells derived from rhesus monkey embryonic stem cells (ESC) had hepatocyte characteristics after the differentiation. METHODS Rhesus monkey ESC were induced towards hepatocyte-like cells via a four-step differentiation process: the formation of embryoid bodies (EB), EB in activin A and insulin-transferrin-selenium medium for 4 days, in fibroblast growth factor (FGF)-4 and bone morphogenetic protein-2 (BMP2) medium for 8 days, in hepatocyte culture medium containing hepatocyte growth factor for 3 days and then with oncostatin M and dexamethasone for another 5 days. Expression of albumin (ALB), glucose-6-phosphatase, α-fetoprotein (AFP) and α-1 antitrypsin (α1-AT) at the mRNA level in differentiated cells were detected by reverse transcription-polymerase chain reaction. The expression of hepatocyte markers AFP, ALB, hepatocyte nuclear factor 4 (HNF4), cytokeratin 8 (CK8), CK19 and cell proliferation marker, Ki67, in the differentiated cells were determined by immunocytochemistry. The ultrastructure of the differentiated cells was examined by electron microscopy. Indocyanine green (ICG) uptake was also explored. RESULTS After induction, some differentiated cells were binucleate, which is typical of hepatocytes. Hepatocyte-specific genes ALB, glucose-6-phosphatase, AFP and α1-AT were expressed in the differentiated cells. The differentiated cells expressed hepatocyte markers AFP, ALB, HNF4, CK8 and CK19 at the protein level. The cells also expressed cell proliferation marker Ki67. Under electron microscopy, the ultrastructures of hepatocyte-like cells, such as mitochondrion and catalase-containing peroxisomes, were observed in the differentiated cells. ICG uptake test was positive in differentiated cells. CONCLUSIONS With cytokine induction, rhesus monkey ESC differentiated into cells displaying morphological features, gene expression patterns and metabolic activities characteristic of hepatocytes.
Collapse
Affiliation(s)
- Xiao Ling Kuai
- Department of Gastroenterology, Affiliated Hospital of Nantong University, School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| | | | | | | | | |
Collapse
|
182
|
Streckfuss-Bömeke K, Jende J, Cheng IF, Hasenfuss G, Guan K. Efficient generation of hepatic cells from multipotent adult mouse germ-line stem cells using an OP9 co-culture system. Cell Reprogram 2013; 16:65-76. [PMID: 24380658 DOI: 10.1089/cell.2013.0057] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
On the basis of their self-renewal capacity and their ability to differentiate into derivatives of all three germ layers, germ line-derived multipotent adult stem cells (maGSCs) from mouse testis might serve as one of preferable sources for pluripotent stem cells in regenerative medicine. In our study, we aimed for an efficient hepatic differentiation protocol that is applicable for both maGSCs and embryonic stem cells (ESCs). We attempted to accomplish this goal by using a new established co-culture system with OP9 stroma cells for direct differentiation of maGSCs and ESCs into hepatic cells. We found that the hepatic differentiation of maGSCs was induced by the OP9 co-culture system in comparison to the gelatin culture. Furthermore, we showed that the combination of OP9 co-culture with activin A resulted in the increased expression of endodermal and early hepatic markers Gata4, Sox17, Foxa2, Hnf4, Afp, and Ttr compared to differentiated cells on gelatin or on OP9 alone. Moreover, the hepatic progenitors were capable of differentiating further into mature hepatic cells, demonstrated by the expression of liver-specific markers Aat, Alb, Tdo2, Krt18, Krt8, Krt19, Cps1, Sek, Cyp7a1, Otc, and Pah. A high percentage of maGSC-derived hepatic progenitors (51% AFP- and 61% DLK1-positive) and mature hepatic-like cells (26% ALB-positive) were achieved using this OP9 co-culture system. These generated hepatic cells successfully demonstrated in vitro functions associated with mature hepatocytes, including albumin and urea secretion, glycogen storage, and uptake of low-density lipoprotein. The established co-culture system for maGSCs into functional hepatic cells might serve as a suitable model to delineate the differentiation process for the generation of high numbers of mature hepatocytes in humans without genetic manipulations and make germ line-derived stem cells a potential autologous and alternative cell source for hepatic transplants in metabolic liver disorders.
Collapse
Affiliation(s)
- Katrin Streckfuss-Bömeke
- 1 Department of Cardiology and Pneumology, Georg-August-University of Göttingen , 37075, Göttingen, Germany
| | | | | | | | | |
Collapse
|
183
|
Szkolnicka D, Farnworth SL, Lucendo-Villarin B, Storck C, Zhou W, Iredale JP, Flint O, Hay DC. Accurate prediction of drug-induced liver injury using stem cell-derived populations. Stem Cells Transl Med 2013; 3:141-8. [PMID: 24375539 DOI: 10.5966/sctm.2013-0146] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Despite major progress in the knowledge and management of human liver injury, there are millions of people suffering from chronic liver disease. Currently, the only cure for end-stage liver disease is orthotopic liver transplantation; however, this approach is severely limited by organ donation. Alternative approaches to restoring liver function have therefore been pursued, including the use of somatic and stem cell populations. Although such approaches are essential in developing scalable treatments, there is also an imperative to develop predictive human systems that more effectively study and/or prevent the onset of liver disease and decompensated organ function. We used a renewable human stem cell resource, from defined genetic backgrounds, and drove them through developmental intermediates to yield highly active, drug-inducible, and predictive human hepatocyte populations. Most importantly, stem cell-derived hepatocytes displayed equivalence to primary adult hepatocytes, following incubation with known hepatotoxins. In summary, we have developed a serum-free, scalable, and shippable cell-based model that faithfully predicts the potential for human liver injury. Such a resource has direct application in human modeling and, in the future, could play an important role in developing renewable cell-based therapies.
Collapse
Affiliation(s)
- Dagmara Szkolnicka
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom; FibromEd Products Ltd., Edinburgh Bio-Quarter, Edinburgh, United Kingdom; Medical Research Council Centre for Inflammation, Edinburgh, United Kingdom; Discovery Toxicology, Bristol-Myers Squibb, Princeton, New Jersey, USA; Department of Oncology, Second Military Medical University, Shanghai Changzheng Hospital, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
184
|
Van Camp JK, Beckers S, Zegers D, Van Hul W. Wnt Signaling and the Control of Human Stem Cell Fate. Stem Cell Rev Rep 2013; 10:207-29. [DOI: 10.1007/s12015-013-9486-8] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
185
|
Hazeltine LB, Selekman JA, Palecek SP. Engineering the human pluripotent stem cell microenvironment to direct cell fate. Biotechnol Adv 2013; 31:1002-19. [PMID: 23510904 PMCID: PMC3758782 DOI: 10.1016/j.biotechadv.2013.03.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 02/20/2013] [Accepted: 03/11/2013] [Indexed: 01/31/2023]
Abstract
Human pluripotent stem cells (hPSCs), including both embryonic stem cells and induced pluripotent stem cells, offer a potential cell source for research, drug screening, and regenerative medicine applications due to their unique ability to self-renew or differentiate to any somatic cell type. Before the full potential of hPSCs can be realized, robust protocols must be developed to direct their fate. Cell fate decisions are based on components of the surrounding microenvironment, including soluble factors, substrate or extracellular matrix, cell-cell interactions, mechanical forces, and 2D or 3D architecture. Depending on their spatio-temporal context, these components can signal hPSCs to either self-renew or differentiate to cell types of the ectoderm, mesoderm, or endoderm. Researchers working at the interface of engineering and biology have identified various factors which can affect hPSC fate, often based on lessons from embryonic development, and they have utilized this information to design in vitro niches which can reproducibly direct hPSC fate. This review highlights culture systems that have been engineered to promote self-renewal or differentiation of hPSCs, with a focus on studies that have elucidated the contributions of specific microenvironmental cues in the context of those culture systems. We propose the use of microsystem technologies for high-throughput screening of spatial-temporal presentation of cues, as this has been demonstrated to be a powerful approach for differentiating hPSCs to desired cell types.
Collapse
Affiliation(s)
| | | | - Sean P. Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin – Madison 1415 Engineering Drive, Madison, WI 53706 USA
| |
Collapse
|
186
|
Costa A, Sarmento B, Seabra V. An evaluation of the latestin vitrotools for drug metabolism studies. Expert Opin Drug Metab Toxicol 2013; 10:103-19. [DOI: 10.1517/17425255.2014.857402] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
187
|
Sun P, Zhou X, Farnworth SL, Patel AH, Hay DC. Modeling human liver biology using stem cell-derived hepatocytes. Int J Mol Sci 2013; 14:22011-21. [PMID: 24201130 PMCID: PMC3856048 DOI: 10.3390/ijms141122011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Revised: 10/28/2013] [Accepted: 10/30/2013] [Indexed: 02/05/2023] Open
Abstract
Stem cell-derived hepatocytes represent promising models to study human liver biology and disease. This concise review discusses the recent progresses in the field, with a focus on human liver disease, drug metabolism and virus infection.
Collapse
Affiliation(s)
- Pingnan Sun
- Shantou University Medical College, Shantou 515041, China; E-Mails: (P.S.); (X.Z.)
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK; E-Mail:
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G11 5JR, UK; E-Mail:
| | - Xiaoling Zhou
- Shantou University Medical College, Shantou 515041, China; E-Mails: (P.S.); (X.Z.)
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK; E-Mail:
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G11 5JR, UK; E-Mail:
| | - Sarah L. Farnworth
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK; E-Mail:
| | - Arvind H. Patel
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G11 5JR, UK; E-Mail:
| | - David C. Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +44-131-651-9549; Fax: +44-131-651-9501
| |
Collapse
|
188
|
Leung A, Nah S, Reid W, Ebata A, Koch C, Monti S, Genereux J, Wiseman R, Wolozin B, Connors L, Berk J, Seldin D, Mostoslavsky G, Kotton D, Murphy G. Induced pluripotent stem cell modeling of multisystemic, hereditary transthyretin amyloidosis. Stem Cell Reports 2013; 1:451-63. [PMID: 24286032 PMCID: PMC3841264 DOI: 10.1016/j.stemcr.2013.10.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 10/04/2013] [Accepted: 10/07/2013] [Indexed: 11/24/2022] Open
Abstract
Familial transthyretin amyloidosis (ATTR) is an autosomal-dominant protein-folding disorder caused by over 100 distinct mutations in the transthyretin (TTR) gene. In ATTR, protein secreted from the liver aggregates and forms fibrils in target organs, chiefly the heart and peripheral nervous system, highlighting the need for a model capable of recapitulating the multisystem complexity of this clinically variable disease. Here, we describe the directed differentiation of ATTR patient-specific iPSCs into hepatocytes that produce mutant TTR, and the cardiomyocytes and neurons normally targeted in the disease. We demonstrate that iPSC-derived neuronal and cardiac cells display oxidative stress and an increased level of cell death when exposed to mutant TTR produced by the patient-matched iPSC-derived hepatocytes, recapitulating essential aspects of the disease in vitro. Furthermore, small molecule stabilizers of TTR show efficacy in this model, validating this iPSC-based, patient-specific in vitro system as a platform for testing therapeutic strategies.
Collapse
Affiliation(s)
- Amy Leung
- Sections of Hematology-Oncology and Computational Biomedicine, Departments of Medicine, Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Center for Regenerative Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | - Shirley K. Nah
- Sections of Hematology-Oncology and Computational Biomedicine, Departments of Medicine, Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Center for Regenerative Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | - Whitney Reid
- Sections of Hematology-Oncology and Computational Biomedicine, Departments of Medicine, Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Center for Regenerative Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | - Atsushi Ebata
- Departments of Pharmacology and Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Clarissa M. Koch
- The Amyloidosis Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Stefano Monti
- Sections of Hematology-Oncology and Computational Biomedicine, Departments of Medicine, Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Joseph C. Genereux
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - R. Luke Wiseman
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Benjamin Wolozin
- Departments of Pharmacology and Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Lawreen H. Connors
- The Amyloidosis Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - John L. Berk
- The Amyloidosis Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - David C. Seldin
- Sections of Hematology-Oncology and Computational Biomedicine, Departments of Medicine, Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- The Amyloidosis Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Gustavo Mostoslavsky
- Center for Regenerative Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | - Darrell N. Kotton
- Center for Regenerative Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | - George J. Murphy
- Sections of Hematology-Oncology and Computational Biomedicine, Departments of Medicine, Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Center for Regenerative Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| |
Collapse
|
189
|
The human constitutive androstane receptor promotes the differentiation and maturation of hepatic-like cells. Dev Biol 2013; 384:155-65. [PMID: 24144921 DOI: 10.1016/j.ydbio.2013.10.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 09/11/2013] [Accepted: 10/12/2013] [Indexed: 11/22/2022]
Abstract
Expression of the constitutive androstane receptor (CAR, NR1I3) is enriched in the mature mammalian liver and increasingly recognized for its prominent role in regulating a myriad of processes including biotransformation, chemical transport, energy metabolism and lipid homeostasis. Previously, we demonstrated that CAR levels were markedly enhanced during the differentiation of hepatic-like cells derived from hESCs, prompting the hypothesis that CAR contributes a key functional role in directing human hepatogenesis. Here we demonstrate that over-expression of CAR in human embryonic stem cells (ESCs), transduced by a lentiviral vector, accelerates the maturation of hepatic-like cells, with CAR over-expressing cells exhibiting a 2.5-fold increase in albumin secretion by day 20 in culture differentiation, and significantly enhanced levels of mRNA expression of several liver-selective markers, including hepatic transcription factors, plasma proteins, biotransformation enzymes, and metabolic enzymes. CAR over-expressing cells also exhibited enhanced CITCO-inducible CYP3A7 enzymatic activity. Knockdown of CAR via siRNA attenuated the differentiation-dependent expression programs. In contrast, expression levels of the pregnane X receptor (PXR), a nuclear receptor most similar to CAR in primary sequence, were negligible in human fetal liver tissues or in the differentiating hESCs, and stable over-expression of PXR in hepatic-induced hESCs failed to enhance expression of hepatic phenotype markers. Together, these results define a novel role for human CAR in hepatic lineage commitment.
Collapse
|
190
|
Vosough M, Omidinia E, Kadivar M, Shokrgozar MA, Pournasr B, Aghdami N, Baharvand H. Generation of functional hepatocyte-like cells from human pluripotent stem cells in a scalable suspension culture. Stem Cells Dev 2013; 22:2693-2705. [PMID: 23731381 DOI: 10.1089/scd.2013.0088] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Recent advances in human embryonic and induced pluripotent stem cell-based therapies in animal models of hepatic failure have led to an increased appreciation of the need to translate the proof-of-principle concepts into more practical and feasible protocols for scale up and manufacturing of functional hepatocytes. In this study, we describe a scalable stirred-suspension bioreactor culture of functional hepatocyte-like cells (HLCs) from the human pluripotent stem cells (hPSCs). To promote the initial differentiation of hPSCs in a carrier-free suspension stirred bioreactor into definitive endoderm, we used rapamycin for "priming" phase and activin A for induction. The cells were further differentiated into HLCs in the same system. HLCs were characterized and then purified based on their physiological function, the uptake of DiI-acetylated low-density lipoprotein (LDL) by flow cytometry without genetic manipulation or antibody labeling. The sorted cells were transplanted into the spleens of mice with acute liver injury from carbon tetrachloride. The differentiated HLCs had multiple features of primary hepatocytes, for example, the expression patterns of liver-specific marker genes, albumin secretion, urea production, collagen synthesis, indocyanin green and LDL uptake, glycogen storage, and inducible cytochrome P450 activity. They increased the survival rate, engrafted successfully into the liver, and continued to present hepatic function (i.e., albumin secretion after implantation). This amenable scaling up and outlined enrichment strategy provides a new platform for generating functional HLCs. This integrated approach may facilitate biomedical applications of the hPSC-derived hepatocytes.
Collapse
Affiliation(s)
- Massoud Vosough
- 1 Department of Biochemistry, Pasteur Institute of Iran , Tehran, Iran
| | | | | | | | | | | | | |
Collapse
|
191
|
Toivonen S, Lundin K, Balboa D, Ustinov J, Tamminen K, Palgi J, Trokovic R, Tuuri T, Otonkoski T. Activin A and Wnt-dependent specification of human definitive endoderm cells. Exp Cell Res 2013; 319:2535-44. [DOI: 10.1016/j.yexcr.2013.07.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 06/28/2013] [Accepted: 07/10/2013] [Indexed: 12/31/2022]
|
192
|
Ogawa S, Surapisitchat J, Virtanen C, Ogawa M, Niapour M, Sugamori KS, Wang S, Tamblyn L, Guillemette C, Hoffmann E, Zhao B, Strom S, Laposa RR, Tyndale RF, Grant DM, Keller G. Three-dimensional culture and cAMP signaling promote the maturation of human pluripotent stem cell-derived hepatocytes. Development 2013; 140:3285-96. [PMID: 23861064 DOI: 10.1242/dev.090266] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Human pluripotent stem cells (hPSCs) represent a novel source of hepatocytes for drug metabolism studies and cell-based therapy for the treatment of liver diseases. These applications are, however, dependent on the ability to generate mature metabolically functional cells from the hPSCs. Reproducible and efficient generation of such cells has been challenging to date, owing to the fact that the regulatory pathways that control hepatocyte maturation are poorly understood. Here, we show that the combination of three-dimensional cell aggregation and cAMP signaling enhance the maturation of hPSC-derived hepatoblasts to a hepatocyte-like population that displays expression profiles and metabolic enzyme levels comparable to those of primary human hepatocytes. Importantly, we also demonstrate that generation of the hepatoblast population capable of responding to cAMP is dependent on appropriate activin/nodal signaling in the definitive endoderm at early stages of differentiation. Together, these findings provide new insights into the pathways that regulate maturation of hPSC-derived hepatocytes and in doing so provide a simple and reproducible approach for generating metabolically functional cell populations.
Collapse
Affiliation(s)
- Shinichiro Ogawa
- McEwen Centre For Regenerative Medicine, University Health Network, Toronto, ON M5G 1L7, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Dianat N, Steichen C, Vallier L, Weber A, Dubart-Kupperschmitt A. Human pluripotent stem cells for modelling human liver diseases and cell therapy. Curr Gene Ther 2013; 13:120-32. [PMID: 23444872 PMCID: PMC3882648 DOI: 10.2174/1566523211313020006] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 01/30/2013] [Accepted: 02/05/2013] [Indexed: 12/24/2022]
Abstract
The liver is affected by many types of diseases, including metabolic disorders and acute liver failure. Orthotopic liver transplantation (OLT) is currently the only effective treatment for life-threatening liver diseases but transplantation of allogeneic hepatocytes has now become an alternative as it is less invasive than OLT and can be performed repeatedly. However, this approach is hampered by the shortage of organ donors, and the problems related to the isolation of high quality adult hepatocytes, their cryopreservation and their absence of proliferation in culture. Liver is also a key organ to assess the pharmacokinetics and toxicology of xenobiotics and for drug discovery, but appropriate cell culture systems are lacking. All these problems have highlighted the need to explore other sources of cells such as stem cells that could be isolated, expanded to yield sufficiently large populations and then induced to differentiate into functional hepatocytes. The presence of a niche of “facultative” progenitor and stem cells in the normal liver has recently been confirmed but they display no telomerase activity. The recent discovery that human induced pluripotent stem cells can be generated from somatic cells has renewed hopes for regenerative medicine and in vitro disease modelling, as these cells are easily accessible. We review here the present progresses, limits and challenges for the generation of functional hepatocytes from human pluripotent stem cells in view of their potential use in regenerative medicine and drug discovery.
Collapse
Affiliation(s)
- Noushin Dianat
- INSERM UMR-S972, Paul Brousse Hospital, Villejuif, F-94807, France
| | | | | | | | | |
Collapse
|
194
|
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: 5.2] [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
|
195
|
Abstract
Differentiation of human embryonic stem (ES) and induced pluripotent stem (iPS) cells into hepatocyte-like cells provides a platform to study the molecular basis of human hepatocyte differentiation, to develop cell culture models of liver disease, and to potentially provide hepatocytes for treatment of end-stage liver disease. Additionally, hepatocyte-like cells generated from human pluripotent stem cells could serve as platforms for drug discovery, determination of pharmaceutical-induced hepatotoxicity, and evaluation of idiosyncratic drug-drug interactions. Here, we describe a step-wise protocol previously developed in our laboratory that facilitates the highly efficient and reproducible differentiation of human pluripotent stem cells into hepatocyte-like cells. Our protocol uses defined culture conditions and closely recapitulates key developmental events that are found to occur during hepatogenesis.
Collapse
Affiliation(s)
- Sunil K Mallanna
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Stephen A Duncan
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
196
|
An in vitro expansion system for generation of human iPS cell-derived hepatic progenitor-like cells exhibiting a bipotent differentiation potential. PLoS One 2013; 8:e67541. [PMID: 23935837 PMCID: PMC3723819 DOI: 10.1371/journal.pone.0067541] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 05/20/2013] [Indexed: 12/15/2022] Open
Abstract
Hepatoblasts, hepatic stem/progenitor cells in liver development, have a high proliferative potential and the ability to differentiate into both hepatocytes and cholangiocytes. In regenerative medicine and drug screening for the treatment of severe liver diseases, human induced pluripotent stem (iPS) cell-derived mature functional hepatocytes are considered to be a potentially good cell source. However, induction of proliferation of these cells is difficult ex vivo. To circumvent this problem, we generated hepatic progenitor-like cells from human iPS cells using serial cytokine treatments in vitro. Highly proliferative hepatic progenitor-like cells were purified by fluorescence-activated cell sorting using antibodies against CD13 and CD133 that are known cell surface markers of hepatic stem/progenitor cells in fetal and adult mouse livers. When the purified CD13highCD133+ cells were cultured at a low density with feeder cells in the presence of suitable growth factors and signaling inhibitors (ALK inhibitor A-83-01 and ROCK inhibitor Y-27632), individual cells gave rise to relatively large colonies. These colonies consisted of two types of cells expressing hepatocytic marker genes (hepatocyte nuclear factor 4α and α-fetoprotein) and a cholangiocytic marker gene (cytokeratin 7), and continued to proliferate over long periods of time. In a spheroid formation assay, these cells were found to express genes required for mature liver function, such as cytochrome P450 enzymes, and secrete albumin. When these cells were cultured in a suitable extracellular matrix gel, they eventually formed a cholangiocytic cyst-like structure with epithelial polarity, suggesting that human iPS cell-derived hepatic progenitor-like cells have a bipotent differentiation ability. Collectively these data indicate that this novel procedure using an in vitro expansion system is useful for not only liver regeneration but also for the determination of molecular mechanisms that regulate liver development.
Collapse
|
197
|
Szkolnicka D, Zhou W, Lucendo-Villarin B, Hay DC. Pluripotent stem cell-derived hepatocytes: potential and challenges in pharmacology. Annu Rev Pharmacol Toxicol 2013; 53:147-59. [PMID: 23294308 DOI: 10.1146/annurev-pharmtox-011112-140306] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The liver is a fascinating organ and performs a wide range of functions necessary for life. Because the hepatocyte is the major functional cell type found in the liver, it is important that we better understand its role in health and disease. Functional hepatocytes have been derived from many sources, including human stem cell populations. These models offer new opportunities to further our understanding of human liver biology from diverse genotypes and, in the future, to facilitate the development of novel medicines or cell-based therapies. This review discusses limitations in current cell-based models and the advantages offered by pluripotent stem cell-derived hepatocytes.
Collapse
Affiliation(s)
- Dagmara Szkolnicka
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | | | | | | |
Collapse
|
198
|
Greenhough S, Bradburn H, Gardner J, Hay DC. Development of an embryoid body-based screening strategy for assessing the hepatocyte differentiation potential of human embryonic stem cells following single-cell dissociation. Cell Reprogram 2013; 15:9-14. [PMID: 23379579 DOI: 10.1089/cell.2012.0049] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We have devised an embryoid body-based screening method for the selection of human embryonic stem cell (hESC) lines capable of forming functional hepatocyte-like cells (HLCs) after single-cell dissociation. The screening method highlighted one cell line from a panel of five that produced albumin-positive cells during embryoid body (EB) formation. Cell lines that did not produce albumin-positive cells during EB formation were shown to respond less well to directed differentiation following single-cell replating. Additionally, the seeding density of the pluripotent populations prior to differentiation was shown to exert a significant effect on the hepatic function of the final population of cells. In summary, we have developed a simple procedure that facilitates the identification of human hESC lines that tolerate single-cell replating and are capable of differentiating to HLCs. Although the hepatic function of cells produced by this method is ∼10-fold lower than our current gold standard stem cell-derived models, we believe that these findings represent an incremental step toward producing HLCs at scale.
Collapse
Affiliation(s)
- Sebastian Greenhough
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh BioQuarter, Scotland
| | | | | | | |
Collapse
|
199
|
Ulvestad M, Nordell P, Asplund A, Rehnström M, Jacobsson S, Holmgren G, Davidson L, Brolén G, Edsbagge J, Björquist P, Küppers-Munther B, Andersson TB. Drug metabolizing enzyme and transporter protein profiles of hepatocytes derived from human embryonic and induced pluripotent stem cells. Biochem Pharmacol 2013; 86:691-702. [PMID: 23856292 DOI: 10.1016/j.bcp.2013.06.029] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 06/26/2013] [Accepted: 06/27/2013] [Indexed: 12/18/2022]
Abstract
Human embryonic and induced pluripotent stem cell-derived hepatocytes (hESC-Hep and hiPSC-Hep) have the potential to provide relevant human in vitro model systems for toxicity testing and drug discovery studies. In this study, the expression and function of important drug metabolizing cytochrome P450 (CYP) enzymes and transporter proteins in hESC-Hep and hiPSC-Hep were compared to cryopreserved human primary hepatocytes (hphep) and HepG2 cells. Overall, CYP activities in hESC-Hep and hiPSC-Hep were much lower than in hphep cultured for 4 h, but CYP1A and 3A activities were comparable to levels in hphep cultured for 48h (CYP1A: 35% and 26% of 48 h hphep, respectively; CYP3A: 80% and 440% of 48 h hphep, respectively). Importantly, in hESC-Hep and hiPSC-Hep, CYP activities were stable or increasing for at least one week in culture which was in contrast to the rapid loss of CYP activities in cultured hphep between 4 and 48 h after plating. With regard to transporters, in hESC-Hep and hiPSC-Hep, pronounced NTCP activity (17% and 29% of 4 h hphep, respectively) and moderate BSEP activity (6% and 8% of 4 h hphep, respectively) were observed. Analyses of mRNA expression and immunocytochemistry supported the observed CYP and transporter activities and showed expression of additional CYPs and transporters. In conclusion, the stable expression and function of CYPs and transporters in hESC-Hep and hiPSC-Hep for at least one week opens up the possibility to reproducibly perform long term and extensive studies, e.g. chronic toxicity testing, in a stem cell-derived hepatic system.
Collapse
Affiliation(s)
- Maria Ulvestad
- DMPK, AstraZeneca R&D Mölndal, Pepparedsleden 1, SE-431 83 Mölndal, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
200
|
The generation of hepatocytes from mesenchymal stem cells and engraftment into the liver. Curr Opin Organ Transplant 2013; 16:69-75. [PMID: 21150616 DOI: 10.1097/mot.0b013e3283424f5b] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Liver transplantation is the ultimate therapeutic option for the treatment of end-stage liver diseases, which, however, is restricted by the shortage of donor organs. Instead hepatocyte transplantation seemed to be a way out, but again marginal donor livers for the isolation of primary human hepatocytes are scarce. The hepatocyte differentiation capacity of mesenchymal stem cells might open a new cell resource to generate hepatocyte-like cells for therapeutical use. RECENT FINDINGS Apart from their potency of hepatocyte differentiation mesenchymal stem cells display pleiotropic biological features including modulation of immunogenicity, anti-inflammatory and anti-apoptotic as well as pro-proliferative impact at the site of tissue or organ lesions. They are mobilized from the bone marrow and migrate to the liver along chemoattractive gradients thus contributing to the humoral and cellular response in tissue repair. The cause of different liver diseases is varying depending on, for example, viral, toxic, nutritional, neoplastic challenges. As known from animal studies mesenchymal stem cells seem to have a beneficial impact on liver regeneration and tissue repair under a variety of liver disease conditions. SUMMARY Their versatile biological features render mesenchymal stem cells an alternate cell resource for the treatment of liver diseases. It is important to know the mechanisms of integration of transplanted cells into the recipient tissue and to understand the communication between donor cells and the host tissue on the molecular level in order to support efficacy of cell transplantation and thus optimize the therapeutical outcome.
Collapse
|