51
|
Wang L, Smith J, Breton C, Clark P, Zhang J, Ying L, Che Y, Lape J, Bell P, Calcedo R, Buza EL, Saveliev A, Bartsevich VV, He Z, White J, Li M, Jantz D, Wilson JM. Meganuclease targeting of PCSK9 in macaque liver leads to stable reduction in serum cholesterol. Nat Biotechnol 2018; 36:717-725. [DOI: 10.1038/nbt.4182] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 06/05/2018] [Indexed: 02/06/2023]
|
52
|
Genetic and Chemical Screenings Identify HDAC3 as a Key Regulator in Hepatic Differentiation of Human Pluripotent Stem Cells. Stem Cell Reports 2018; 11:22-31. [PMID: 29861165 PMCID: PMC6066908 DOI: 10.1016/j.stemcr.2018.05.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 12/17/2022] Open
Abstract
Hepatocyte-like cells (HLCs) derived from human pluripotent stem cells (hPSCs) offer a promising cell resource for disease modeling and transplantation. However, differentiated HLCs exhibit an immature phenotype and comprise a heterogeneous population. Thus, a better understanding of HLC differentiation will improve the likelihood of future application. Here, by taking advantage of CRISPR-Cas9-based genome-wide screening technology and a high-throughput hPSC screening platform with a reporter readout, we identified several potential genetic regulators of HLC differentiation. By using a chemical screening approach within our platform, we also identified compounds that can further promote HLC differentiation and preserve the characteristics of in vitro cultured primary hepatocytes. Remarkably, both screenings identified histone deacetylase 3 (HDAC3) as a key regulator in hepatic differentiation. Mechanistically, HDAC3 formed a complex with liver transcriptional factors, e.g., HNF4, and co-regulated the transcriptional program during hepatic differentiation. This study highlights a broadly useful approach for studying and optimizing hPSC differentiation. Genome-wide genetic screening to identify regulators in hepatic differentiation HDAC3 regulates hepatic differentiation Small molecule CI-994 treatment improves hepatic differentiation A broadly useful approach for studying and the optimization of hPSC differentiation
Collapse
|
53
|
Sambathkumar R, Akkerman R, Dastidar S, Roelandt P, Kumar M, Bajaj M, Mestre Rosa AR, Helsen N, Vanslembrouck V, Kalo E, Khurana S, Laureys J, Gysemans C, Faas MM, de Vos P, Verfaillie CM. Generation of hepatocyte- and endocrine pancreatic-like cells from human induced endodermal progenitor cells. PLoS One 2018; 13:e0197046. [PMID: 29750821 PMCID: PMC5947914 DOI: 10.1371/journal.pone.0197046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 04/25/2018] [Indexed: 01/27/2023] Open
Abstract
Multipotent Adult Progenitor Cells (MAPCs) are one potential stem cell source to generate functional hepatocytes or β-cells. However, human MAPCs have less plasticity than pluripotent stem cells (PSCs), as their ability to generate endodermal cells is not robust. Here we studied the role of 14 transcription factors (TFs) in reprogramming MAPCs to induced endodermal progenitor cells (iENDO cells), defined as cells that can be long-term expanded and differentiated to both hepatocyte- and endocrine pancreatic-like cells. We demonstrated that 14 TF-iENDO cells can be expanded for at least 20 passages, differentiate spontaneously to hepatocyte-, endocrine pancreatic-, gut tube-like cells as well as endodermal tumor formation when grafted in immunodeficient mice. Furthermore, iENDO cells can be differentiated in vitro into hepatocyte- and endocrine pancreatic-like cells. However, the pluripotency TF OCT4, which is not silenced in iENDO cells, may contribute to the incomplete differentiation to mature cells in vitro and to endodermal tumor formation in vivo. Nevertheless, the studies presented here provide evidence that reprogramming of adult stem cells to an endodermal intermediate progenitor, which can be expanded and differentiate to multiple endodermal cell types, might be a valid alternative for the use of PSCs for creation of endodermal cell types.
Collapse
Affiliation(s)
- Rangarajan Sambathkumar
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
- * E-mail: (CMV); (RS)
| | - Renate Akkerman
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
- University of Groningen, University Medical Center Groningen (UMCG), Pathology and Medical Biology, Division of Medical Biology, Section Immunoendocrinology, Groningen, The Netherlands
| | - Sumitava Dastidar
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Philip Roelandt
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Manoj Kumar
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Manmohan Bajaj
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Ana Rita Mestre Rosa
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Nicky Helsen
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Veerle Vanslembrouck
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Eric Kalo
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Satish Khurana
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Jos Laureys
- KU Leuven, Department of Clinical and Experimental Medicine, Clinical and Experimental Endocrinology unit, Leuven, Belgium
| | - Conny Gysemans
- KU Leuven, Department of Clinical and Experimental Medicine, Clinical and Experimental Endocrinology unit, Leuven, Belgium
| | - Marijke M. Faas
- University of Groningen, University Medical Center Groningen (UMCG), Pathology and Medical Biology, Division of Medical Biology, Section Immunoendocrinology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen (UMCG), Department of Obstetrics and Gynecology, Groningen, The Netherlands
| | - Paul de Vos
- University of Groningen, University Medical Center Groningen (UMCG), Pathology and Medical Biology, Division of Medical Biology, Section Immunoendocrinology, Groningen, The Netherlands
| | - Catherine M. Verfaillie
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
- * E-mail: (CMV); (RS)
| |
Collapse
|
54
|
Zhang RR, Koido M, Tadokoro T, Ouchi R, Matsuno T, Ueno Y, Sekine K, Takebe T, Taniguchi H. Human iPSC-Derived Posterior Gut Progenitors Are Expandable and Capable of Forming Gut and Liver Organoids. Stem Cell Reports 2018; 10:780-793. [PMID: 29429958 PMCID: PMC5919071 DOI: 10.1016/j.stemcr.2018.01.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/10/2018] [Accepted: 01/11/2018] [Indexed: 02/06/2023] Open
Abstract
Early endoderm progenitors naturally possess robust propagating potential to develop a majority of meter-long gastrointestinal tracts and are therefore considered as a promising source for therapy. Here, we demonstrated the reproducible generation of human CDX2+ posterior gut endoderm cells (PGECs) from five induced pluripotent stem cell clones by manipulating FGF, TGF, and WNT signaling. Transcriptome analysis suggested that putative PGECs harbored an intermediate signature profile between definitive endoderm and organ-specific endoderm. We found that combinatorial EGF, VEGF, FGF2, Chir99021, and A83-01 treatments selectively amplify storable PGECs up to 1021 cell scale without any gene transduction or feeder use. PGECs, compared with induced pluripotent stem cells, showed stable differentiation propensity into multiple endodermal lineages without teratoma formation. Furthermore, transplantation of PGEC-derived liver bud organoids showed therapeutic potential against fulminant liver failure. Together, the robustly amplified PGECs may be a promising cellular source for endoderm-derived organoids in studying human development, modeling disease, and, ultimately, therapy. Successful activation of posteriorization program using human iPSC-derived endoderm CDX2+ PGECs exhibit a robust amplification potential after passaging and freezing PGECs with differentiation potential into hindgut and liver bud organoids PGEC liver bud transplant is therapeutically effective for acute liver failure
Collapse
Affiliation(s)
- Ran-Ran Zhang
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan; Division of Gastroenterology, Hepatology & Nutrition, Developmental Biology, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA
| | - Masaru Koido
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Tomomi Tadokoro
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Rie Ouchi
- Division of Gastroenterology, Hepatology & Nutrition, Developmental Biology, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA
| | - Tatsuya Matsuno
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Yasuharu Ueno
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Keisuke Sekine
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Takanori Takebe
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan; Division of Gastroenterology, Hepatology & Nutrition, Developmental Biology, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA; Advanced Medical Research Center, Yokohama City University, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan; PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan; Division of Gastroenterology, Hepatology & Nutrition, Developmental Biology, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA.
| |
Collapse
|
55
|
Trott J, Tan EK, Ong S, Titmarsh DM, Denil SLIJ, Giam M, Wong CK, Wang J, Shboul M, Eio M, Cooper-White J, Cool SM, Rancati G, Stanton LW, Reversade B, Dunn NR. Long-Term Culture of Self-renewing Pancreatic Progenitors Derived from Human Pluripotent Stem Cells. Stem Cell Reports 2018; 8:1675-1688. [PMID: 28591650 PMCID: PMC5470345 DOI: 10.1016/j.stemcr.2017.05.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/15/2017] [Accepted: 05/15/2017] [Indexed: 12/21/2022] Open
Abstract
Pluripotent stem cells have been proposed as an unlimited source of pancreatic β cells for studying and treating diabetes. However, the long, multi-step differentiation protocols used to generate functional β cells inevitably exhibit considerable variability, particularly when applied to pluripotent cells from diverse genetic backgrounds. We have developed culture conditions that support long-term self-renewal of human multipotent pancreatic progenitors, which are developmentally more proximal to the specialized cells of the adult pancreas. These cultured pancreatic progenitor (cPP) cells express key pancreatic transcription factors, including PDX1 and SOX9, and exhibit transcriptomes closely related to their in vivo counterparts. Upon exposure to differentiation cues, cPP cells give rise to pancreatic endocrine, acinar, and ductal lineages, indicating multilineage potency. Furthermore, cPP cells generate insulin+ β-like cells in vitro and in vivo, suggesting that they offer a convenient alternative to pluripotent cells as a source of adult cell types for modeling pancreatic development and diabetes. Culture on 3T3 cells enables long-term self-renewal of human pancreatic progenitors Proliferation requires EGF, FGF10, retinoic acid, and inhibition of Notch and TGF-β Cultured progenitors upregulate genes required for mitosis and telomere maintenance Pancreatic duct and β-like cells are generated in vitro and in vivo
Collapse
Affiliation(s)
- Jamie Trott
- Institute of Medical Biology, Agency for Science Technology and Research (A(∗)STAR), 8a Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore.
| | - Ee Kim Tan
- Institute of Medical Biology, Agency for Science Technology and Research (A(∗)STAR), 8a Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Sheena Ong
- Institute of Medical Biology, Agency for Science Technology and Research (A(∗)STAR), 8a Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Drew M Titmarsh
- Institute of Medical Biology, Agency for Science Technology and Research (A(∗)STAR), 8a Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Simon L I J Denil
- Institute of Medical Biology, Agency for Science Technology and Research (A(∗)STAR), 8a Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Maybelline Giam
- Institute of Medical Biology, Agency for Science Technology and Research (A(∗)STAR), 8a Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Cheng Kit Wong
- Institute of Medical Biology, Agency for Science Technology and Research (A(∗)STAR), 8a Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Jiaxu Wang
- Genome Institute of Singapore, Agency for Science Technology and Research (A(∗)STAR), 60 Biopolis Street, #02-01, Singapore 138672, Singapore
| | - Mohammad Shboul
- Institute of Medical Biology, Agency for Science Technology and Research (A(∗)STAR), 8a Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Michelle Eio
- Institute of Medical Biology, Agency for Science Technology and Research (A(∗)STAR), 8a Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Justin Cooper-White
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Simon M Cool
- Institute of Medical Biology, Agency for Science Technology and Research (A(∗)STAR), 8a Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Giulia Rancati
- Institute of Medical Biology, Agency for Science Technology and Research (A(∗)STAR), 8a Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Lawrence W Stanton
- Genome Institute of Singapore, Agency for Science Technology and Research (A(∗)STAR), 60 Biopolis Street, #02-01, Singapore 138672, Singapore
| | - Bruno Reversade
- Institute of Medical Biology, Agency for Science Technology and Research (A(∗)STAR), 8a Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - N Ray Dunn
- Institute of Medical Biology, Agency for Science Technology and Research (A(∗)STAR), 8a Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
56
|
Petersen MB, Gonçalves CA, Kim YH, Grapin-Botton A. Recapitulating and Deciphering Human Pancreas Development From Human Pluripotent Stem Cells in a Dish. Curr Top Dev Biol 2018; 129:143-190. [DOI: 10.1016/bs.ctdb.2018.02.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
57
|
Ameri J, Borup R, Prawiro C, Ramond C, Schachter KA, Scharfmann R, Semb H. Efficient Generation of Glucose-Responsive Beta Cells from Isolated GP2 + Human Pancreatic Progenitors. Cell Rep 2017; 19:36-49. [PMID: 28380361 DOI: 10.1016/j.celrep.2017.03.032] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 02/10/2017] [Accepted: 03/09/2017] [Indexed: 12/29/2022] Open
Abstract
Stem cell-based therapy for type 1 diabetes would benefit from implementation of a cell purification step at the pancreatic endoderm stage. This would increase the safety of the final cell product, allow the establishment of an intermediate-stage stem cell bank, and provide a means for upscaling β cell manufacturing. Comparative gene expression analysis revealed glycoprotein 2 (GP2) as a specific cell surface marker for isolating pancreatic endoderm cells (PECs) from differentiated hESCs and human fetal pancreas. Isolated GP2+ PECs efficiently differentiated into glucose responsive insulin-producing cells in vitro. We found that in vitro PEC proliferation declines due to enhanced expression of the cyclin-dependent kinase (CDK) inhibitors CDKN1A and CDKN2A. However, we identified a time window when reducing CDKN1A or CDKN2A expression increased proliferation and yield of GP2+ PECs. Altogether, our results contribute tools and concepts toward the isolation and use of PECs as a source for the safe production of hPSC-derived β cells.
Collapse
Affiliation(s)
- Jacqueline Ameri
- The Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark; Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, BMC, B10, 22184 Lund, Sweden
| | - Rehannah Borup
- Center for Genomic Medicine, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Christy Prawiro
- The Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Cyrille Ramond
- INSERM U1016, University Paris-Descartes, Cochin Institute, 75014 Paris, France
| | - Karen A Schachter
- The Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Raphael Scharfmann
- INSERM U1016, University Paris-Descartes, Cochin Institute, 75014 Paris, France
| | - Henrik Semb
- The Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark; Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, BMC, B10, 22184 Lund, Sweden.
| |
Collapse
|
58
|
Gao Y, Zhang X, Zhang L, Cen J, Ni X, Liao X, Yang C, Li Y, Chen X, Zhang Z, Shu Y, Cheng X, Hay DC, Lai D, Pan G, Wei G, Hui L. Distinct Gene Expression and Epigenetic Signatures in Hepatocyte-like Cells Produced by Different Strategies from the Same Donor. Stem Cell Reports 2017; 9:1813-1824. [PMID: 29173899 PMCID: PMC5785700 DOI: 10.1016/j.stemcr.2017.10.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/22/2017] [Accepted: 10/23/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatocyte-like cells (HLCs) can be generated through directed differentiation or transdifferentiation. Employing two strategies, we generated induced pluripotent stem cell (iPSC)-HLCs and hiHeps from the same donor cell line. Both types of HLCs clustered distinctly from each other during gene expression profiling. In particular, differences existed in gene expression for phase II drug metabolism and lipid accumulation, underpinned by H3K27 acetylation status in iPSC-HLCs and hiHeps. While distinct phenotypes were achieved in vitro, both types of HLCs demonstrated similar phenotypes following transplantation into Fah-deficient mice. In conclusion, functional HLCs can be obtained from the same donor using two strategies. Global gene expression defined the differences between those populations in vitro. Importantly, both HLCs displayed partial but markedly improved hepatic function following transplantation in vivo, demonstrating plasticity and the potential for cell-based modeling in the dish and cell-based therapy in the future. hiHeps and iPSC-HLCs from the same donor are compared hiHeps and iPSC-HLCs show distinct expression patterns and hepatic functions Different expressions in hiHeps and iPSC-HLCs are partially attributed to H3K27ac Both HLCs are further matured in the in vivo microenvironment of livers
Collapse
Affiliation(s)
- Yimeng Gao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoran Zhang
- CAS Key Laboratory of Computational Biology, Collaborative Innovation Center for Genetics and Developmental Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ludi Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jin Cen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xuan Ni
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Xiaoying Liao
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Chenxi Yang
- State Key Laboratory of Bioreactor Engineering, School of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
| | - Ying Li
- CAS Key Laboratory of Computational Biology, Collaborative Innovation Center for Genetics and Developmental Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaotao Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhao Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yajing Shu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xin Cheng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - David C Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Dongmei Lai
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200030, China
| | - Guoyu Pan
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Gang Wei
- CAS Key Laboratory of Computational Biology, Collaborative Innovation Center for Genetics and Developmental Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Lijian Hui
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
59
|
Ortmann D, Vallier L. Variability of human pluripotent stem cell lines. Curr Opin Genet Dev 2017; 46:179-185. [DOI: 10.1016/j.gde.2017.07.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/07/2017] [Accepted: 07/14/2017] [Indexed: 12/23/2022]
|
60
|
Cao S, Yu S, Chen Y, Wang X, Zhou C, Liu Y, Kuang J, Liu H, Li D, Ye J, Qin Y, Chu S, Wu L, Guo L, Li Y, Shu X, Chen J, Liu J, Pei D. Chemical reprogramming of mouse embryonic and adult fibroblast into endoderm lineage. J Biol Chem 2017; 292:19122-19132. [PMID: 28935668 DOI: 10.1074/jbc.m117.812537] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/19/2017] [Indexed: 12/21/2022] Open
Abstract
We report here an approach to redirecting somatic cell fate under chemically defined conditions without transcription factors. We start by converting mouse embryonic fibroblasts to epithelial-like cells with chemicals and growth factors. Subsequent cell fate mapping reveals a robust induction of SOX17 in the resulting epithelial-like cells that can be further reprogrammed to endodermal progenitor cells. Interestingly, these cells can self-renew in vitro and further differentiate into albumin-producing hepatocytes that can rescue mice from acute liver injury. Our results demonstrate a rational approach to convert mouse embryonic fibroblasts to hepatocytes and suggest that this mechanism-driven approach may be generalized for other cells.
Collapse
Affiliation(s)
- Shangtao Cao
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,the University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Shengyong Yu
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yan Chen
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xiaoshan Wang
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,the University of the Chinese Academy of Sciences, Beijing 100049, China.,the Guangzhou Branch of the Supercomputing Center of the Chinese Academy of Sciences, Guangzhou 510530, China, and
| | - Chunhua Zhou
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,the University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Yuting Liu
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Junqi Kuang
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,the University of the Chinese Academy of Sciences, Beijing 100049, China
| | - He Liu
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,the University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Dongwei Li
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,the University of the Chinese Academy of Sciences, Beijing 100049, China.,the Guangzhou Branch of the Supercomputing Center of the Chinese Academy of Sciences, Guangzhou 510530, China, and
| | - Jing Ye
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yue Qin
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,the University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Shilong Chu
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Linlin Wu
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,the University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Guo
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yinxiong Li
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xiaodong Shu
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Jiekai Chen
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China.,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,the Guangzhou Branch of the Supercomputing Center of the Chinese Academy of Sciences, Guangzhou 510530, China, and
| | - Jing Liu
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China, .,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Duanqing Pei
- From the Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Beijing 100049, China, .,the Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,the Guangzhou Branch of the Supercomputing Center of the Chinese Academy of Sciences, Guangzhou 510530, China, and
| |
Collapse
|
61
|
Peng G, Tam PPL, Jing N. Lineage specification of early embryos and embryonic stem cells at the dawn of enabling technologies. Natl Sci Rev 2017. [DOI: 10.1093/nsr/nwx093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Abstract
Establishment of progenitor cell populations and lineage diversity during embryogenesis and the differentiation of pluripotent stem cells is a fascinating and intricate biological process. Conceptually, an understanding of this developmental process provides a framework to integrate stem-cell pluripotency, cell competence and differentiating potential with the activity of extrinsic and intrinsic molecular determinants. The recent advent of enabling technologies of high-resolution transcriptome analysis at the cellular, population and spatial levels proffers the capability of gaining deeper insights into the attributes of the gene regulatory network and molecular signaling in lineage specification and differentiation. In this review, we provide a snapshot of the emerging enabling genomic technologies that contribute to the study of development and stem-cell biology.
Collapse
Affiliation(s)
- Guangdun Peng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Patrick P. L. Tam
- Embryology Unit, Children's Medical Research Institute, School of Medical Sciences, Sydney Medical School, University of Sydney, NSW 2145, Australia
| | - Naihe Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
62
|
Footprint-free human fetal foreskin derived iPSCs: A tool for modeling hepatogenesis associated gene regulatory networks. Sci Rep 2017; 7:6294. [PMID: 28740077 PMCID: PMC5524812 DOI: 10.1038/s41598-017-06546-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 06/13/2017] [Indexed: 12/17/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are similar to embryonic stem cells and can be generated from somatic cells. We have generated episomal plasmid-based and integration-free iPSCs (E-iPSCs) from human fetal foreskin fibroblast cells (HFF1). We used an E-iPSC-line to model hepatogenesis in vitro. The HLCs were characterized biochemically, i.e. glycogen storage, ICG uptake and release, UREA and bile acid production, as well as CYP3A4 activity. Ultra-structure analysis by electron microscopy revealed the presence of lipid and glycogen storage, tight junctions and bile canaliculi- all typical features of hepatocytes. Furthermore, the transcriptome of undifferentiated E-iPSC, DE, HE and HLCs were compared to that of fetal liver and primary human hepatocytes (PHH). K-means clustering identified 100 clusters which include developmental stage-specific groups of genes, e.g. OCT4 expression at the undifferentiated stage, SOX17 marking the DE stage, DLK and HNF6 the HE stage, HNF4α and Albumin is specific to HLCs, fetal liver and adult liver (PHH) stage. We use E-iPSCs for modeling gene regulatory networks associated with human hepatogenesis and gastrulation in general.
Collapse
|
63
|
Jacobson EF, Tzanakakis ES. Human pluripotent stem cell differentiation to functional pancreatic cells for diabetes therapies: Innovations, challenges and future directions. J Biol Eng 2017; 11:21. [PMID: 28680477 PMCID: PMC5494890 DOI: 10.1186/s13036-017-0066-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 06/02/2017] [Indexed: 12/18/2022] Open
Abstract
Recent advances in the expansion and directed pancreatogenic differentiation of human pluripotent stem cells (hPSCs) have intensified efforts to generate functional pancreatic islet cells, especially insulin-secreting β-cells, for cell therapies against diabetes. However, the consistent generation of glucose-responsive insulin-releasing cells remains challenging. In this article, we first present basic concepts of pancreatic organogenesis, which frequently serves as a basis for engineering differentiation regimens. Next, past and current efforts are critically discussed for the conversion of hPSCs along pancreatic cell lineages, including endocrine β-cells and α-cells, as well as exocrine cells with emphasis placed on the later stages of commitment. Finally, major challenges and future directions are examined, such as the identification of factors for in vivo maturation, large-scale culture and post processing systems, cell loss during differentiation, culture economics, efficiency, and efficacy and exosomes and miRNAs in pancreatic differentiation.
Collapse
Affiliation(s)
- Elena F Jacobson
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby St., Room 276A, Medford, MA 02155 USA
| | - Emmanuel S Tzanakakis
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby St., Room 276A, Medford, MA 02155 USA.,Tufts Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA 02111 USA
| |
Collapse
|
64
|
Coskun E, Ercin M, Gezginci‐Oktayoglu S. The Role of Epigenetic Regulation and Pluripotency‐Related MicroRNAs in Differentiation of Pancreatic Stem Cells to Beta Cells. J Cell Biochem 2017; 119:455-467. [DOI: 10.1002/jcb.26203] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/08/2017] [Indexed: 01/17/2023]
Affiliation(s)
- Ediz Coskun
- Faculty of ScienceBiology DepartmentMolecular Biology Section, Istanbul UniversityVezneciler 34134IstanbulTurkey
| | - Merve Ercin
- Faculty of ScienceBiology DepartmentMolecular Biology Section, Istanbul UniversityVezneciler 34134IstanbulTurkey
| | - Selda Gezginci‐Oktayoglu
- Faculty of ScienceBiology DepartmentMolecular Biology Section, Istanbul UniversityVezneciler 34134IstanbulTurkey
| |
Collapse
|
65
|
Ma X, Zhu S. Chemical strategies for pancreatic β cell differentiation, reprogramming, and regeneration. Acta Biochim Biophys Sin (Shanghai) 2017; 49:289-301. [PMID: 28338772 DOI: 10.1093/abbs/gmx008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Indexed: 12/13/2022] Open
Abstract
Generation of unlimited functional pancreatic β cells is critical for the study of pancreatic biology and treatment of diabetes mellitus. Recent advances have suggested several promising directions, including directed differentiation of pancreatic β cells from pluripotent stem cells, reprogramming of pancreatic β cells from other types of somatic cells, and stimulated proliferation and enhanced functions of existing pancreatic β cells. Small molecules are useful in generating unlimited numbers of functional pancreatic cells in vitro and could be further developed as drugs to stimulate endogenous pancreatic regeneration. Here, we provide an updated summary of recent major achievements in pancreatic β cell differentiation, reprogramming, proliferation, and function. These studies will eventually lead to significant advances in the field of pancreatic biology and regeneration.
Collapse
Affiliation(s)
- Xiaojie Ma
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Saiyong Zhu
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
66
|
Peng G, Jing N. The genome-wide molecular regulation of mouse gastrulation embryo. SCIENCE CHINA-LIFE SCIENCES 2017; 60:363-369. [PMID: 28251461 DOI: 10.1007/s11427-016-0285-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/25/2016] [Indexed: 11/24/2022]
Abstract
The diverse morphologies among vertebrate species stems from the evolution of a basic body plan that is constituted by a spatially organized ensemble of tissue lineage progenitors. At gastrulation, this body plan is established through a coordinated morphogenetic process and the delineation of tissue lineages that are driven by the activity of the genome. To explore the molecular mechanisms, in a comprehensive context, it is imperative to glean an understanding of the region- and population-specific genetic activity underpinning this fundamental developmental process. In this review, we outline the recent progresses and the future directions in studies of genome activity for the regulation of mouse embryogenesis at gastrulation.
Collapse
Affiliation(s)
- Guangdun Peng
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Naihe Jing
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
67
|
Jiang FX, Li K, Archer M, Mehta M, Jamieson E, Charles A, Dickinson JE, Matsumoto M, Morahan G. Differentiation of Islet Progenitors Regulated by Nicotinamide into Transcriptome-Verified β Cells That Ameliorate Diabetes. Stem Cells 2017; 35:1341-1354. [DOI: 10.1002/stem.2567] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 11/21/2016] [Accepted: 12/14/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Fang-Xu Jiang
- Islet Cell Development Program, Harry Perkins Institute of Medical Research, and Centre for Medical Research
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, and Centre for Medical Research
| | - Kevin Li
- Islet Cell Development Program, Harry Perkins Institute of Medical Research, and Centre for Medical Research
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, and Centre for Medical Research
| | | | - Munish Mehta
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, and Centre for Medical Research
| | - Emma Jamieson
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, and Centre for Medical Research
| | - Adrian Charles
- School of Women's and Infants' Health; The University of Western Australia; Nedlands Australia
| | - Jan E. Dickinson
- School of Women's and Infants' Health; The University of Western Australia; Nedlands Australia
| | | | - Grant Morahan
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, and Centre for Medical Research
| |
Collapse
|
68
|
Tiyaboonchai A, Cardenas-Diaz FL, Ying L, Maguire JA, Sim X, Jobaliya C, Gagne AL, Kishore S, Stanescu DE, Hughes N, De Leon DD, French DL, Gadue P. GATA6 Plays an Important Role in the Induction of Human Definitive Endoderm, Development of the Pancreas, and Functionality of Pancreatic β Cells. Stem Cell Reports 2017; 8:589-604. [PMID: 28196690 PMCID: PMC5355564 DOI: 10.1016/j.stemcr.2016.12.026] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 12/20/2022] Open
Abstract
Induced pluripotent stem cells were created from a pancreas agenesis patient with a mutation in GATA6. Using genome-editing technology, additional stem cell lines with mutations in both GATA6 alleles were generated and demonstrated a severe block in definitive endoderm induction, which could be rescued by re-expression of several different GATA family members. Using the endodermal progenitor stem cell culture system to bypass the developmental block at the endoderm stage, cell lines with mutations in one or both GATA6 alleles could be differentiated into β-like cells but with reduced efficiency. Use of suboptimal doses of retinoic acid during pancreas specification revealed a more severe phenotype, more closely mimicking the patient’s disease. GATA6 mutant β-like cells fail to secrete insulin upon glucose stimulation and demonstrate defective insulin processing. These data show that GATA6 plays a critical role in endoderm and pancreas specification and β-like cell functionality in humans. GATA6 is required for definitive endoderm specification in human ES/iPS cells Bypassing the endoderm defect allows GATA6 mutants to generate β-like cells Suboptimal retinoic acid signaling blocks pancreas specification in GATA6 mutants GATA6 is critical for human β cell function in vitro
Collapse
Affiliation(s)
- Amita Tiyaboonchai
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, CTRB 5012, 3501 Civic Center Boulevard, Philadelphia, PA 19104, USA; Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fabian L Cardenas-Diaz
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, CTRB 5012, 3501 Civic Center Boulevard, Philadelphia, PA 19104, USA; Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lei Ying
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, CTRB 5012, 3501 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Jean Ann Maguire
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, CTRB 5012, 3501 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Xiuli Sim
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, CTRB 5012, 3501 Civic Center Boulevard, Philadelphia, PA 19104, USA; Graduate Program in Cell and Molecular Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chintan Jobaliya
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, CTRB 5012, 3501 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Alyssa L Gagne
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, CTRB 5012, 3501 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Siddharth Kishore
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, CTRB 5012, 3501 Civic Center Boulevard, Philadelphia, PA 19104, USA; Graduate Program in Cell and Molecular Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Diana E Stanescu
- Division of Endocrinology, Department of Pediatrics, Perelman School of Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nkecha Hughes
- Clinical and Translational Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Diva D De Leon
- Division of Endocrinology, Department of Pediatrics, Perelman School of Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Deborah L French
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, CTRB 5012, 3501 Civic Center Boulevard, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paul Gadue
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, CTRB 5012, 3501 Civic Center Boulevard, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
69
|
Higuchi A, Suresh Kumar S, Ling QD, Alarfaj AA, Munusamy MA, Murugan K, Hsu ST, Benelli G, Umezawa A. Polymeric design of cell culture materials that guide the differentiation of human pluripotent stem cells. Prog Polym Sci 2017. [DOI: 10.1016/j.progpolymsci.2016.09.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
70
|
Willmer T, Cooper A, Peres J, Omar R, Prince S. The T-Box transcription factor 3 in development and cancer. Biosci Trends 2017; 11:254-266. [DOI: 10.5582/bst.2017.01043] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Tarryn Willmer
- Department of Human Biology, Faculty of Health Sciences, Anzio Road, University of Cape Town
| | - Aretha Cooper
- Department of Human Biology, Faculty of Health Sciences, Anzio Road, University of Cape Town
| | - Jade Peres
- Department of Human Biology, Faculty of Health Sciences, Anzio Road, University of Cape Town
| | - Rehana Omar
- Department of Human Biology, Faculty of Health Sciences, Anzio Road, University of Cape Town
| | - Sharon Prince
- Department of Human Biology, Faculty of Health Sciences, Anzio Road, University of Cape Town
| |
Collapse
|
71
|
Rajaei B, Shamsara M, Amirabad LM, Massumi M, Sanati MH. Pancreatic Endoderm-Derived From Diabetic Patient-Specific Induced Pluripotent Stem Cell Generates Glucose-Responsive Insulin-Secreting Cells. J Cell Physiol 2016; 232:2616-2625. [DOI: 10.1002/jcp.25459] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/14/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Bahareh Rajaei
- Department of Medical Genetics; Institute of Medical Biotechnology; National Institute of Genetic Engineering and Biotechnology; Tehran Iran
| | - Mehdi Shamsara
- National Center for Transgenic Mouse Research; Institute of Agricultural Biotechnology; National Institute of Genetic Engineering and Biotechnology; Tehran Iran
| | - Leila Mohammadi Amirabad
- National Center for Transgenic Mouse Research; Institute of Agricultural Biotechnology; National Institute of Genetic Engineering and Biotechnology; Tehran Iran
- School of Advanced Technologies in Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Mohammad Massumi
- Department of Physiology; University of Toronto; Toronto Ontario Canada
- Lunenfeld-Tanenbaum Research Institute; Mount Sinai Hospital; Toronto Ontario Canada
| | - Mohammad Hossein Sanati
- Department of Medical Genetics; Institute of Medical Biotechnology; National Institute of Genetic Engineering and Biotechnology; Tehran Iran
| |
Collapse
|
72
|
Luo X, Wang H, Leighton J, O'Sullivan M, Wang P. Generation of endoderm lineages from pluripotent stem cells. Regen Med 2016; 12:77-89. [PMID: 27976977 DOI: 10.2217/rme-2016-0086] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Definitive endoderm is the cellular precursor to respiratory- and digestive-related organs such as lungs, stomach, liver, pancreas and intestine. Endodermal lineage cells derived from pluripotent stem cells (PSCs) in vitro are a potentially unlimited resource for regenerative medicine. These cells are useful tools for studying the physiology, pathogenesis and medical therapies involving these tissues, and great progress has been achieved in PSCs differentiation protocols. In this review, we will focus on the most common and/or advanced differentiation strategies currently used in generating endodermal lineage cells from PSCs. A brief discussion about the effect of early definitive endoderm differentiation on the final development products will follow.
Collapse
Affiliation(s)
- Xie Luo
- Department of Cellular & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Han Wang
- Department of Cellular & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jake Leighton
- Department of Cellular & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Mara O'Sullivan
- Department of Cellular & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Pei Wang
- Department of Cellular & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
73
|
Bulk cell density and Wnt/TGFbeta signalling regulate mesendodermal patterning of human pluripotent stem cells. Nat Commun 2016; 7:13602. [PMID: 27934856 PMCID: PMC5155150 DOI: 10.1038/ncomms13602] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 10/17/2016] [Indexed: 12/22/2022] Open
Abstract
In vitro differentiation of human pluripotent stem cells (hPSCs) recapitulates early aspects of human embryogenesis, but the underlying processes are poorly understood and controlled. Here we show that modulating the bulk cell density (BCD: cell number per culture volume) deterministically alters anteroposterior patterning of primitive streak (PS)-like priming. The BCD in conjunction with the chemical WNT pathway activator CHIR99021 results in distinct paracrine microenvironments codifying hPSCs towards definitive endoderm, precardiac or presomitic mesoderm within the first 24 h of differentiation, respectively. Global gene expression and secretome analysis reveals that TGFß superfamily members, antagonist of Nodal signalling LEFTY1 and CER1, are paracrine determinants restricting PS progression. These data result in a tangible model disclosing how hPSC-released factors deflect CHIR99021-induced lineage commitment over time. By demonstrating a decisive, functional role of the BCD, we show its utility as a method to control lineage-specific differentiation. Furthermore, these findings have profound consequences for inter-experimental comparability, reproducibility, bioprocess optimization and scale-up.
Collapse
|
74
|
Al Madhoun A, Ali H, AlKandari S, Atizado VL, Akhter N, Al-Mulla F, Atari M. Defined three-dimensional culture conditions mediate efficient induction of definitive endoderm lineage from human umbilical cord Wharton's jelly mesenchymal stem cells. Stem Cell Res Ther 2016; 7:165. [PMID: 27852316 PMCID: PMC5111269 DOI: 10.1186/s13287-016-0426-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 10/18/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) are gaining increasing interest as an alternative source of stem cells for regenerative medicine applications. Definitive endoderm (DE) specification is a prerequisite for the development of vital organs such as liver and pancreas. Hence, efficient induction of the DE lineage from stem cells is crucial for subsequent generation of clinically relevant cell types. Here we present a defined 3D differentiation protocol of WJ-MSCs into DE cells. METHODS WJ-MSCs were cultured in suspension to generate spheroids, about 1500 cells each, for 7 days. The serum-free differentiation media contained specific growth factors, cytokines, and small molecules that specifically regulate signaling pathways including sonic hedgehog, bone morphogenetic protein, Activin/Wnt, and Notch. RESULTS We obtained more than 85 % DE cells as shown with FACS analysis using antibodies directed against the DE marker CXCR4. In addition, biochemical and molecular analysis of bona-fide DE markers revealed a time-course induction of Sox17, CXCR4, and FoxA2. Focused PCR-based array also indicated a specific induction into the DE lineage. CONCLUSIONS In this study, we report an efficient serum-free protocol to differentiate WJ-MSCs into DE cells utilizing 3D spheroid formation. Our approach might aid in the development of new protocols to obtain DE-derivative lineages including liver-like and pancreatic insulin-producing cells.
Collapse
Affiliation(s)
| | - Hamad Ali
- Research Division, Dasman Diabetes Institute, 1180 Dasman, Kuwait
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center, Kuwait University, Al-Jabriya, Kuwait
| | - Sarah AlKandari
- Research Division, Dasman Diabetes Institute, 1180 Dasman, Kuwait
| | | | - Nadeem Akhter
- Research Division, Dasman Diabetes Institute, 1180 Dasman, Kuwait
| | - Fahd Al-Mulla
- Department of Pathology, Molecular Pathology Unit, Faculty of Medicine, Health Sciences Center, Kuwait University, Al-Jabriya, Kuwait
| | - Maher Atari
- UIC Regenerative Medicine Research Institute, International University of Catalonia, Barcelona, Spain
| |
Collapse
|
75
|
Raju R, Chau D, Cho DS, Park Y, Verfaillie CM, Hu WS. Cell Expansion During Directed Differentiation of Stem Cells Toward the Hepatic Lineage. Stem Cells Dev 2016; 26:274-284. [PMID: 27806669 DOI: 10.1089/scd.2016.0119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The differentiation of human pluripotent stem cells toward the hepatocyte lineage can potentially provide an unlimited source of functional hepatocytes for transplantation and extracorporeal bioartificial liver applications. It is anticipated that the quantities of cells needed for these applications will be in the order of 109-1010 cells, because of the size of the liver. An ideal differentiation protocol would be to enable directed differentiation to the hepatocyte lineage with simultaneous cell expansion. We introduced a cell expansion stage after the commitment of human embryonic stem cells to the endodermal lineage, to allow for at least an eightfold increase in cell number, with continuation of cell maturation toward the hepatocyte lineage. The progressive changes in the transcriptome were measured by expression array, and the expression dynamics of certain lineage markers was measured by mass cytometry during the differentiation and expansion process. The findings revealed that while cells were expanding they were also capable of progressing in their differentiation toward the hepatocyte lineage. In addition, our transcriptome, protein and functional studies, including albumin secretion, drug-induced CYP450 expression and urea production, all indicated that the hepatocyte-like cells obtained with or without cell expansion are very similar. This method of simultaneous cell expansion and hepatocyte differentiation should facilitate obtaining large quantities of cells for liver cell applications.
Collapse
Affiliation(s)
- Ravali Raju
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| | - David Chau
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota.,3 Department of Biomedical Engineering, University of Minnesota , Minneapolis, Minnesota
| | - Dong Seong Cho
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| | - Yonsil Park
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| | - Catherine M Verfaillie
- 4 Department of Development and Regeneration, Stem Cell Institute Leuven , KU Leuven, Leuven, Belgium
| | - Wei-Shou Hu
- 1 Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota.,2 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|
76
|
Takayama K, Mizuguchi H. Generation of human pluripotent stem cell-derived hepatocyte-like cells for drug toxicity screening. Drug Metab Pharmacokinet 2016; 32:12-20. [PMID: 28012798 DOI: 10.1016/j.dmpk.2016.10.408] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 10/03/2016] [Accepted: 10/17/2016] [Indexed: 01/22/2023]
Abstract
Because drug-induced liver injury is one of the main reasons for drug development failures, it is important to perform drug toxicity screening in the early phase of pharmaceutical development. Currently, primary human hepatocytes are most widely used for the prediction of drug-induced liver injury. However, the sources of primary human hepatocytes are limited, making it difficult to supply the abundant quantities required for large-scale drug toxicity screening. Therefore, there is an urgent need for a novel unlimited, efficient, inexpensive, and predictive model which can be applied for large-scale drug toxicity screening. Human embryonic stem (ES) cells and induced pluripotent stem (iPS) cells are able to replicate indefinitely and differentiate into most of the body's cell types, including hepatocytes. It is expected that hepatocyte-like cells generated from human ES/iPS cells (human ES/iPS-HLCs) will be a useful tool for drug toxicity screening. To apply human ES/iPS-HLCs to various applications including drug toxicity screening, homogenous and functional HLCs must be differentiated from human ES/iPS cells. In this review, we will introduce the current status of hepatocyte differentiation technology from human ES/iPS cells and a novel method to predict drug-induced liver injury using human ES/iPS-HLCs.
Collapse
Affiliation(s)
- Kazuo Takayama
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan; The Keihanshin Consortium for Fostering the Next Generation of Global Leaders in Research (K-CONNEX), Kyoto University, Kyoto 606-8302, Japan; PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan; Laboratory of Hepatocyte Regulation, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan.
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan; Laboratory of Hepatocyte Regulation, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
77
|
Calderon D, Bardot E, Dubois N. Probing early heart development to instruct stem cell differentiation strategies. Dev Dyn 2016; 245:1130-1144. [PMID: 27580352 DOI: 10.1002/dvdy.24441] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 08/20/2016] [Accepted: 08/20/2016] [Indexed: 12/19/2022] Open
Abstract
Scientists have studied organs and their development for centuries and, along that path, described models and mechanisms explaining the developmental principles of organogenesis. In particular, with respect to the heart, new fundamental discoveries are reported continuously that keep changing the way we think about early cardiac development. These discoveries are driven by the need to answer long-standing questions regarding the origin of the earliest cells specified to the cardiac lineage, the differentiation potential of distinct cardiac progenitor cells, and, very importantly, the molecular mechanisms underlying these specification events. As evidenced by numerous examples, the wealth of developmental knowledge collected over the years has had an invaluable impact on establishing efficient strategies to generate cardiovascular cell types ex vivo, from either pluripotent stem cells or via direct reprogramming approaches. The ability to generate functional cardiovascular cells in an efficient and reliable manner will contribute to therapeutic strategies aimed at alleviating the increasing burden of cardiovascular disease and morbidity. Here we will discuss the recent discoveries in the field of cardiac progenitor biology and their translation to the pluripotent stem cell model to illustrate how developmental concepts have instructed regenerative model systems in the past and promise to do so in the future. Developmental Dynamics 245:1130-1144, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Damelys Calderon
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Evan Bardot
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Nicole Dubois
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| |
Collapse
|
78
|
Zhang X, Chen Y, Ye Y, Wang J, Wang H, Yuan G, Lin Z, Wu Y, Zhang Y, Lin X. Wnt signaling promotes hindgut fate commitment through regulating multi-lineage genes during hESC differentiation. Cell Signal 2016; 29:12-22. [PMID: 27693749 DOI: 10.1016/j.cellsig.2016.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 09/22/2016] [Accepted: 09/27/2016] [Indexed: 12/22/2022]
Abstract
Wnt signaling plays essential roles in both embryonic pattern formation and postembryonic tissue homoestasis. High levels of Wnt activity repress foregut identity and facilitate hindgut fate through forming a gradient of Wnt signaling activity along the anterior-posterior axis. Here, we examined the mechanisms of Wnt signaling in hindgut development by differentiating human embryonic stem cells (hESCs) into the hindgut progenitors. We observed severe morphological changes when Wnt signaling was blocked by using Wnt antagonist Dkk1. We performed deep-transcriptome sequencing (RNA-seq) and identified 240 Wnt-activated genes and 2023 Wnt-repressed genes, respectively. Clusters of Wnt targets showed enrichment in specific biological functions, such as "gastrointestinal or skeletal development" in the Wnt-activated targets and "neural or immune system development" in the Wnt-repressed targets. Moreover, we adopted a high-throughput chromatin immunoprecipitation and deep sequencing (ChIP-seq) approach to identify the genomic regions through which Wnt-activated transcription factor TCF7L2 regulated transcription. We identified 83 Wnt direct target candidates, including the hindgut marker CDX2 and the genes relevant to morphogenesis (MSX1, MSX2, LEF1, T, PDGFRB etc.) through combinatorial analysis of the RNA-seq and ChIP-seq data. Together, our study identified a series of direct and indirect Wnt targets in hindgut differentiation, and uncovered the diverse mechanisms of Wnt signaling in regulating multi-lineage differentiation.
Collapse
Affiliation(s)
- Xiujuan Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ying Chen
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ying Ye
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jianfeng Wang
- Core Genomic Facility, CAS Key Laboratory of Genome Sciences & Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Hong Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Guohong Yuan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhe Lin
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yihui Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xinhua Lin
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Division of Developmental Biology, Cincinnati Childrens Hospital Medical Center, Cincinnati, OH, United States; State Key Laboratory of Genetic Engineering, Institute of Genetics, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
79
|
Wang Y, Qin J, Wang S, Zhang W, Duan J, Zhang J, Wang X, Yan F, Chang M, Liu X, Feng B, Liu J, Pei X. Conversion of Human Gastric Epithelial Cells to Multipotent Endodermal Progenitors using Defined Small Molecules. Cell Stem Cell 2016; 19:449-461. [PMID: 27452176 DOI: 10.1016/j.stem.2016.06.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 02/18/2016] [Accepted: 06/13/2016] [Indexed: 12/30/2022]
Abstract
Endodermal stem/progenitor cells have diverse potential applications in research and regenerative medicine, so a readily available source could have widespread uses. Here we describe derivation of human induced endodermal progenitor cells (hiEndoPCs) from gastrointestinal epithelial cells using a cocktail of defined small molecules along with support from tissue-specific mesenchymal feeders. The hiEndoPCs show clonal expansion in culture and give rise to hepatocytes, pancreatic endocrine cells, and intestinal epithelial cells when treated with defined soluble molecules directing differentiation. The hiEndoPC-derived hepatocytes are able to rescue liver failure in Fah-/-Rag2-/- mice after transplantation, and, unlike hESCs, transplanted hiEndoPCs do not give rise to teratomas. Since human gastric epithelial cells are readily available from donors of many ages, this conversion strategy can generate clonally expandable cell populations with a variety of potential applications, including personalized drug screening and therapeutic strategies for liver failure and diabetes.
Collapse
Affiliation(s)
- Yunfang Wang
- Tissue Engineering Lab, Beijing Institute of Transfusion Medicine, Beijing 100850, China.
| | - Jinhua Qin
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing 100850, China; South China Research Center for Stem Cell and Regenerative Medicine, South China Institute of Biomedicine, Guangzhou 510005, China
| | - Shuyong Wang
- Tissue Engineering Lab, Beijing Institute of Transfusion Medicine, Beijing 100850, China; Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing 100850, China; South China Research Center for Stem Cell and Regenerative Medicine, South China Institute of Biomedicine, Guangzhou 510005, China
| | - Wencheng Zhang
- Tissue Engineering Lab, Beijing Institute of Transfusion Medicine, Beijing 100850, China
| | - Jialei Duan
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jing Zhang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xin Wang
- Key Laboratory of National Education, Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Huhhot 010000, China; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Fang Yan
- Tissue Engineering Lab, Beijing Institute of Transfusion Medicine, Beijing 100850, China
| | - Mingyang Chang
- Tissue Engineering Lab, Beijing Institute of Transfusion Medicine, Beijing 100850, China
| | - Xiaofang Liu
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing 100850, China
| | - Bo Feng
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jiang Liu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xuetao Pei
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing 100850, China; South China Research Center for Stem Cell and Regenerative Medicine, South China Institute of Biomedicine, Guangzhou 510005, China.
| |
Collapse
|
80
|
Toivonen S, Malinen MM, Küblbeck J, Petsalo A, Urtti A, Honkakoski P, Otonkoski T. Regulation of Human Pluripotent Stem Cell-Derived Hepatic Cell Phenotype by Three-Dimensional Hydrogel Models. Tissue Eng Part A 2016; 22:971-84. [PMID: 27329070 DOI: 10.1089/ten.tea.2016.0127] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Human-induced pluripotent stem cell (hiPSC)-derived hepatocytes are anticipated as important surrogates for primary human hepatocytes in applications ranging from basic research to drug discovery and regenerative medicine. Although methods for differentiating hepatocyte-like cells (HLCs) from hiPSCs have developed remarkably, the limited yield of fully functional HLCs is still a major obstacle to their utility. A three-dimensional (3D) culture environment could improve the in vitro hepatic maturation of HLCs. Here we compare 3D hydrogel models of hiPSC-derived HLCs in agarose microwells (3D Petri Dish; 3DPD), nanofibrillar cellulose hydrogels (Growdex; 3DNFC), or animal extracellular matrix-based hydrogels (3D Matrigel; 3DMG). In all the tested 3D biomaterial systems, HLCs formed aggregates. In comparison with two-dimensional monolayer culture, 3DPD and 3DMG models showed both phenotypic and functional enhancement in HLCs over 2.5 weeks of 3D culture. Specifically, we found higher hepatocyte-specific gene expression levels and enhanced cytochrome P450 functions. Our work suggests that transferring HLCs into 3D hydrogel systems can expedite the hepatic maturation of HLCs irrespective of the biochemical nature of the 3D hydrogel. Both plant-based nonembedding and animal-based embedding 3D hydrogel models enhanced the maturation.
Collapse
Affiliation(s)
- Sanna Toivonen
- 1 Research Programs Unit, Molecular Neurology, Biomedicum Stem Cell Center, University of Helsinki , Helsinki, Finland
| | - Melina M Malinen
- 2 Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki , Helsinki, Finland
| | - Jenni Küblbeck
- 3 School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland & Biocenter Kuopio , Kuopio, Finland
| | - Aleksanteri Petsalo
- 3 School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland & Biocenter Kuopio , Kuopio, Finland
| | - Arto Urtti
- 2 Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki , Helsinki, Finland .,3 School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland & Biocenter Kuopio , Kuopio, Finland
| | - Paavo Honkakoski
- 3 School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland & Biocenter Kuopio , Kuopio, Finland
| | - Timo Otonkoski
- 1 Research Programs Unit, Molecular Neurology, Biomedicum Stem Cell Center, University of Helsinki , Helsinki, Finland .,4 Children's Hospital, Helsinki University Central Hospital , Helsinki, Finland
| |
Collapse
|
81
|
Devito L, Jacquet L, Petrova A, Miere C, Wood V, Kadeva N, Cornwell G, Codognotto S, Stephenson E, Ilic D. Generation of KCL034 clinical grade human embryonic stem cell line. Stem Cell Res 2016; 16:184-8. [PMID: 27345810 PMCID: PMC4757774 DOI: 10.1016/j.scr.2015.12.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 12/29/2015] [Indexed: 11/21/2022] Open
Abstract
The KCL034 human embryonic stem cell line was derived from a normal healthy blastocyst donated for research. The ICM was isolated using laser microsurgery and plated on γ-irradiated human foreskin fibroblasts. Both the derivation and cell line propagation were performed in an animal product-free environment and under current Good Manufacturing Practice (cGMP) standards. Pluripotent state and differentiation potential were confirmed by in vitro assays. The line was also validated for sterility, specific and non-specific human pathogens.
Collapse
Affiliation(s)
- Liani Devito
- Stem Cell Laboratories, Division of Women's Health, Faculty of Life Sciences and Medicine, King's College London and Assisted Conception Unit, Guys' Hospital, London, United Kingdom
| | - Laureen Jacquet
- Stem Cell Laboratories, Division of Women's Health, Faculty of Life Sciences and Medicine, King's College London and Assisted Conception Unit, Guys' Hospital, London, United Kingdom
| | - Anastasia Petrova
- Stem Cell Laboratories, Division of Women's Health, Faculty of Life Sciences and Medicine, King's College London and Assisted Conception Unit, Guys' Hospital, London, United Kingdom
| | - Cristian Miere
- Stem Cell Laboratories, Division of Women's Health, Faculty of Life Sciences and Medicine, King's College London and Assisted Conception Unit, Guys' Hospital, London, United Kingdom
| | - Victoria Wood
- Stem Cell Laboratories, Division of Women's Health, Faculty of Life Sciences and Medicine, King's College London and Assisted Conception Unit, Guys' Hospital, London, United Kingdom
| | - Neli Kadeva
- Stem Cell Laboratories, Division of Women's Health, Faculty of Life Sciences and Medicine, King's College London and Assisted Conception Unit, Guys' Hospital, London, United Kingdom
| | - Glenda Cornwell
- Stem Cell Laboratories, Division of Women's Health, Faculty of Life Sciences and Medicine, King's College London and Assisted Conception Unit, Guys' Hospital, London, United Kingdom
| | - Stefano Codognotto
- Stem Cell Laboratories, Division of Women's Health, Faculty of Life Sciences and Medicine, King's College London and Assisted Conception Unit, Guys' Hospital, London, United Kingdom
| | - Emma Stephenson
- Stem Cell Laboratories, Division of Women's Health, Faculty of Life Sciences and Medicine, King's College London and Assisted Conception Unit, Guys' Hospital, London, United Kingdom
| | - Dusko Ilic
- Stem Cell Laboratories, Division of Women's Health, Faculty of Life Sciences and Medicine, King's College London and Assisted Conception Unit, Guys' Hospital, London, United Kingdom.
| |
Collapse
|
82
|
Szkolnicka D, Hay DC. Concise Review: Advances in Generating Hepatocytes from Pluripotent Stem Cells for Translational Medicine. Stem Cells 2016; 34:1421-6. [PMID: 27015786 PMCID: PMC4982058 DOI: 10.1002/stem.2368] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/24/2016] [Accepted: 03/10/2016] [Indexed: 12/23/2022]
Abstract
The liver is one of the major organs in the human body. Severe or prolonged exposure of the liver to different factors may cause life-threatening disease, which necessitates donor organ transplantation. While orthotopic liver transplantation can be used to effectively treat liver failure, it is an invasive procedure, which is severely limited by organ donation. Therefore, alternative sources of liver support have been proposed and studied. This includes the use of pluripotent stem cell-derived hepatocytes as a renewable source of cells for therapy. In addition to cell-based therapies, in vitro engineered liver tissue provides powerful models for human drug discovery and disease modeling. This review focuses on the generation of hepatocyte-like cells from pluripotent stem cells and their application in translational medicine. Stem Cells 2016;34:1421-1426.
Collapse
Affiliation(s)
- Dagmara Szkolnicka
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - David C Hay
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| |
Collapse
|
83
|
lncRHOXF1, a Long Noncoding RNA from the X Chromosome That Suppresses Viral Response Genes during Development of the Early Human Placenta. Mol Cell Biol 2016; 36:1764-75. [PMID: 27066803 DOI: 10.1128/mcb.01098-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/07/2016] [Indexed: 11/20/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) can regulate gene expression in a cell-specific fashion during development. Here, we identify a novel lncRNA from the X chromosome that we named lncRHOXF1 and which is abundantly expressed in trophectoderm and primitive endoderm cells of human blastocyst-stage embryos. lncRHOXF1 is a spliced and polyadenylated lncRNA about 1 kb in length that is found in both the nuclear and cytoplasmic compartments of in vitro differentiated human trophectoderm progenitor cells. Gain-of-function experiments in human embryonic stem cells, which normally lack lncRHOXF1 RNA, revealed that lncRHOXF1 reduced proliferation and favored cellular differentiation. lncRHOXF1 knockdown using small interfering RNAs (siRNAs) in human trophectoderm progenitors increased expression of viral response genes, including type I interferon. Sendai virus infection of human trophectoderm progenitor cells increased lncRHOXF1 RNA levels, and siRNA-mediated disruption of lncRHOXF1 during infection reduced the expression of viral response genes leading to higher virus replication. Thus, lncRHOXF1 RNA is the first example of a lncRNA that regulates the host response to viral infections in human placental progenitor cells, and we propose that it functions as a repressor of the viral response during early human development.
Collapse
|
84
|
Jiang FX, Morahan G. Insulin-secreting β cells require a post-genomic concept. World J Diabetes 2016; 7:198-208. [PMID: 27226815 PMCID: PMC4873311 DOI: 10.4239/wjd.v7.i10.198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 03/18/2016] [Indexed: 02/05/2023] Open
Abstract
Pancreatic insulin-secreting β cells are essential in maintaining normal glucose homeostasis accomplished by highly specialized transcription of insulin gene, of which occupies up to 40% their transcriptome. Deficiency of these cells causes diabetes mellitus, a global public health problem. Although tremendous endeavors have been made to generate insulin-secreting cells from human pluripotent stem cells (i.e., primitive cells capable of giving rise to all cell types in the body), a regenerative therapy to diabetes has not yet been established. Furthermore, the nomenclature of β cells has become inconsistent, confusing and controversial due to the lack of standardized positive controls of developmental stage-matched in vivo cells. In order to minimize this negative impact and facilitate critical research in this field, a post-genomic concept of pancreatic β cells might be helpful. In this review article, we will briefly describe how β cells were discovered and islet lineage is developed that may help understand the cause of nomenclatural controversy, suggest a post-genomic definition and finally provide a conclusive remark on future research of this pivotal cell.
Collapse
|
85
|
Emechebe U, Kumar P P, Rozenberg JM, Moore B, Firment A, Mirshahi T, Moon AM. T-box3 is a ciliary protein and regulates stability of the Gli3 transcription factor to control digit number. eLife 2016; 5. [PMID: 27046536 PMCID: PMC4829432 DOI: 10.7554/elife.07897] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 03/05/2016] [Indexed: 12/17/2022] Open
Abstract
Crucial roles for T-box3 in development are evident by severe limb malformations and other birth defects caused by T-box3 mutations in humans. Mechanisms whereby T-box3 regulates limb development are poorly understood. We discovered requirements for T-box at multiple stages of mouse limb development and distinct molecular functions in different tissue compartments. Early loss of T-box3 disrupts limb initiation, causing limb defects that phenocopy Sonic Hedgehog (Shh) mutants. Later ablation of T-box3 in posterior limb mesenchyme causes digit loss. In contrast, loss of anterior T-box3 results in preaxial polydactyly, as seen with dysfunction of primary cilia or Gli3-repressor. Remarkably, T-box3 is present in primary cilia where it colocalizes with Gli3. T-box3 interacts with Kif7 and is required for normal stoichiometry and function of a Kif7/Sufu complex that regulates Gli3 stability and processing. Thus, T-box3 controls digit number upstream of Shh-dependent (posterior mesenchyme) and Shh-independent, cilium-based (anterior mesenchyme) Hedgehog pathway function. DOI:http://dx.doi.org/10.7554/eLife.07897.001 Mutations in the gene that encodes a protein called T-box3 cause serious birth defects, including deformities of the hands and feet, via poorly understood mechanisms. Several other proteins are also important for ensuring that limbs develop correctly. These include the Sonic Hedgehog protein, which controls a signaling pathway that determines whether a protein called Gli3 is converted into its “repressor” form. The hair-like structures called primary cilia that sit on the surface of animal cells also contain Gli3, and processes within these structures control the production of the Gli3-repressor. Emechebe, Kumar et al. have now studied genetically engineered mice in which the production of the T-box3 protein was stopped at different stages of mouse development. This revealed that turning off T-box3 production early in development causes many parts of the limb not to form. This type of defect appears to be the same as that seen in mice that lack the Sonic Hedgehog protein. If the production of T-box3 is turned off later in mouse development in the rear portion of the developing limb, the limb starts to develop but doesn’t develop enough rear toes. When T-box3 production is turned off in the front portion of the developing limbs, mice are born with too many front toes. This latter problem mimics the effects seen in mice that are unable to produce Gli3-repressor or that have defective primary cilia. Further investigation unexpectedly revealed that T-box3 is found in primary cilia and localizes to the same regions of the cilia as the Gli3-repressor. Furthermore, T-box3 also interacts with a protein complex that controls the stability of Gli3 and processes it into the Gli3-repressor form. In the future, it will be important to determine how T-box3 controls the stability of Gli3 and whether that process occurs in the primary cilia or in other parts of the cell where T-box3 and Gli3 coexist, such as the nucleus. This could help us understand how T-box3 and Sonic Hedgehog signaling contribute to other aspects of development and to certain types of cancer. DOI:http://dx.doi.org/10.7554/eLife.07897.002
Collapse
Affiliation(s)
- Uchenna Emechebe
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, United States
| | - Pavan Kumar P
- Weis Center for Research, Geisinger Clinic, Danville, United States
| | | | - Bryn Moore
- Weis Center for Research, Geisinger Clinic, Danville, United States
| | - Ashley Firment
- Weis Center for Research, Geisinger Clinic, Danville, United States
| | - Tooraj Mirshahi
- Weis Center for Research, Geisinger Clinic, Danville, United States
| | - Anne M Moon
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, United States.,Weis Center for Research, Geisinger Clinic, Danville, United States.,Department of Human Genetics, University of Utah, Salt Lake City, United States.,Department of Pediatrics, University of Utah, Salt Lake City, United States
| |
Collapse
|
86
|
Endothelin-1 supports clonal derivation and expansion of cardiovascular progenitors derived from human embryonic stem cells. Nat Commun 2016; 7:10774. [PMID: 26952167 PMCID: PMC4786749 DOI: 10.1038/ncomms10774] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 01/19/2016] [Indexed: 01/10/2023] Open
Abstract
Coronary arteriogenesis is a central step in cardiogenesis, requiring coordinated generation and integration of endothelial cell and vascular smooth muscle cells. At present, it is unclear whether the cell fate programme of cardiac progenitors to generate complex muscular or vascular structures is entirely cell autonomous. Here we demonstrate the intrinsic ability of vascular progenitors to develop and self-organize into cardiac tissues by clonally isolating and expanding second heart field cardiovascular progenitors using WNT3A and endothelin-1 (EDN1) human recombinant proteins. Progenitor clones undergo long-term expansion and differentiate primarily into endothelial and smooth muscle cell lineages in vitro, and contribute extensively to coronary-like vessels in vivo, forming a functional human–mouse chimeric circulatory system. Our study identifies EDN1 as a key factor towards the generation and clonal derivation of ISL1+ vascular intermediates, and demonstrates the intrinsic cell-autonomous nature of these progenitors to differentiate and self-organize into functional vasculatures in vivo. Understanding coronary vessels development provides basis for regenerative strategies. Here, Soh et al. identify endothelin-1 as a key molecule driving long-term expansion of ISL1+ bipotent vascular progenitors derived from human embryonic stem cells, and show that these cells can regenerate coronary vessels in mice.
Collapse
|
87
|
Techniques of Human Embryonic Stem Cell and Induced Pluripotent Stem Cell Derivation. Arch Immunol Ther Exp (Warsz) 2016; 64:349-70. [PMID: 26939778 PMCID: PMC5021740 DOI: 10.1007/s00005-016-0385-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 11/17/2015] [Indexed: 12/22/2022]
Abstract
Developing procedures for the derivation of human pluripotent stem cells (PSCs) gave rise to novel pathways into regenerative medicine research. For many years, stem cells have attracted attention as a potentially unlimited cell source for cellular therapy in neurodegenerative disorders, cardiovascular diseases, and spinal cord injuries, for example. In these studies, adult stem cells were insufficient; therefore, many attempts were made to obtain PSCs by other means. This review discusses key issues concerning the techniques of pluripotent cell acquisition. Technical and ethical issues hindered the medical use of somatic cell nuclear transfer and embryonic stem cells. Therefore, induced PSCs (iPSCs) emerged as a powerful technique with great potential for clinical applications, patient-specific disease modelling and pharmaceutical studies. The replacement of viral vectors or the administration of analogous proteins or chemical compounds during cell reprogramming are modifications designed to reduce tumorigenesis risk and to augment the procedure efficiency. Intensified analysis of new PSC lines revealed other barriers to overcome, such as epigenetic memory, disparity between human and mouse pluripotency, and variable response to differentiation of some iPSC lines. Thus, multidimensional verification must be conducted to fulfil strict clinical-grade requirements. Nevertheless, the first clinical trials in patients with spinal cord injury and macular dystrophy were recently carried out with differentiated iPSCs, encouraging alternative strategies for potential autologous cellular therapies.
Collapse
|
88
|
Mohammadnia A, Yaqubi M, Pourasgari F, Neely E, Fallahi H, Massumi M. Signaling and Gene Regulatory Networks Governing Definitive Endoderm Derivation From Pluripotent Stem Cells. J Cell Physiol 2016; 231:1994-2006. [PMID: 26755186 DOI: 10.1002/jcp.25308] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 01/06/2016] [Indexed: 11/07/2022]
Abstract
The generation of definitive endoderm (DE) from pluripotent stem cells (PSCs) is a fundamental stage in the formation of highly organized visceral organs, such as the liver and pancreas. Currently, there is a need for a comprehensive study that illustrates the involvement of different signaling pathways and their interactions in the derivation of DE cells from PSCs. This study aimed to identify signaling pathways that have the greatest influence on DE formation using analyses of transcriptional profiles, protein-protein interactions, protein-DNA interactions, and protein localization data. Using this approach, signaling networks involved in DE formation were constructed using systems biology and data mining tools, and the validity of the predicted networks was confirmed experimentally by measuring the mRNA levels of hub genes in several PSCs-derived DE cell lines. Based on our analyses, seven signaling pathways, including the BMP, ERK1-ERK2, FGF, TGF-beta, MAPK, Wnt, and PIP signaling pathways and their interactions, were found to play a role in the derivation of DE cells from PSCs. Lastly, the core gene regulatory network governing this differentiation process was constructed. The results of this study could improve our understanding surrounding the efficient generation of DE cells for the regeneration of visceral organs. J. Cell. Physiol. 231: 1994-2006, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Abdulshakour Mohammadnia
- Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Moein Yaqubi
- Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.,Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, Quebec, Canada.,Douglas Mental Health University Institute, McGill University, Montréal, Quebec, Canada
| | - Farzaneh Pourasgari
- Department of Biotechnology, Razi Vaccine and Serum Research Institute, Karaj, Iran.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Eric Neely
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Hossein Fallahi
- Department of Biology, School of Science, Razi University, Kermanshah, Iran.,Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Massumi
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| |
Collapse
|
89
|
El-Badawy A, El-Badri N. The cell cycle as a brake for β-cell regeneration from embryonic stem cells. Stem Cell Res Ther 2016; 7:9. [PMID: 26759123 PMCID: PMC4711007 DOI: 10.1186/s13287-015-0274-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The generation of insulin-producing β cells from stem cells in vitro provides a promising source of cells for cell transplantation therapy in diabetes. However, insulin-producing cells generated from human stem cells show deficiency in many functional characteristics compared with pancreatic β cells. Recent reports have shown molecular ties between the cell cycle and the differentiation mechanism of embryonic stem (ES) cells, assuming that cell fate decisions are controlled by the cell cycle machinery. Both β cells and ES cells possess unique cell cycle machinery yet with significant contrasts. In this review, we compare the cell cycle control mechanisms in both ES cells and β cells, and highlight the fundamental differences between pluripotent cells of embryonic origin and differentiated β cells. Through critical analysis of the differences of the cell cycle between these two cell types, we propose that the cell cycle of ES cells may act as a brake for β-cell regeneration. Based on these differences, we discuss the potential of modulating the cell cycle of ES cells for the large-scale generation of functionally mature β cells in vitro. Further understanding of the factors that modulate the ES cell cycle will lead to new approaches to enhance the production of functional mature insulin-producing cells, and yield a reliable system to generate bona fide β cells in vitro.
Collapse
Affiliation(s)
- Ahmed El-Badawy
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Sheikh Zayed District, 12588, 6th of October City, Giza, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Sheikh Zayed District, 12588, 6th of October City, Giza, Egypt.
| |
Collapse
|
90
|
Human pancreatic beta-like cells converted from fibroblasts. Nat Commun 2016; 7:10080. [PMID: 26733021 PMCID: PMC4729817 DOI: 10.1038/ncomms10080] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 11/02/2015] [Indexed: 02/06/2023] Open
Abstract
Pancreatic beta cells are of great interest for biomedical research and regenerative medicine. Here we show the conversion of human fibroblasts towards an endodermal cell fate by employing non-integrative episomal reprogramming factors in combination with specific growth factors and chemical compounds. On initial culture, converted definitive endodermal progenitor cells (cDE cells) are specified into posterior foregut-like progenitor cells (cPF cells). The cPF cells and their derivatives, pancreatic endodermal progenitor cells (cPE cells), can be greatly expanded. A screening approach identified chemical compounds that promote the differentiation and maturation of cPE cells into functional pancreatic beta-like cells (cPB cells) in vitro. Transplanted cPB cells exhibit glucose-stimulated insulin secretion in vivo and protect mice from chemically induced diabetes. In summary, our study has important implications for future strategies aimed at generating high numbers of functional beta cells, which may help restoring normoglycemia in patients suffering from diabetes.
Collapse
|
91
|
Jiang FX, Morahan G. Multipotent pancreas progenitors: Inconclusive but pivotal topic. World J Stem Cells 2015; 7:1251-1261. [PMID: 26730269 PMCID: PMC4691693 DOI: 10.4252/wjsc.v7.i11.1251] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/20/2015] [Accepted: 11/11/2015] [Indexed: 02/07/2023] Open
Abstract
The establishment of multipotent pancreas progenitors (MPP) should have a significant impact not only on the ontology of the pancreas, but also for the translational research of glucose-responding endocrine β-cells. Deficiency of the latter may lead to the pandemic type 1 or type 2 diabetes mellitus, a metabolic disorder. An ideal treatment of which would potentially be the replacement of destroyed or failed β-cells, by restoring function of endogenous pancreatic endocrine cells or by transplantation of donor islets or in vitro generated insulin-secreting cells. Thus, considerable research efforts have been devoted to identify MPP candidates in the pre- and post-natal pancreas for the endogenous neogenesis or regeneration of endocrine insulin-secreting cells. In order to advance this inconclusive but critical field, we here review the emerging concepts, recent literature and newest developments of potential MPP and propose measures that would assist its forward progression.
Collapse
|
92
|
Pluripotency Factors on Their Lineage Move. Stem Cells Int 2015; 2016:6838253. [PMID: 26770212 PMCID: PMC4684880 DOI: 10.1155/2016/6838253] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 07/30/2015] [Accepted: 08/03/2015] [Indexed: 12/19/2022] Open
Abstract
Pluripotent stem cells are characterised by continuous self-renewal while maintaining the potential to differentiate into cells of all three germ layers. Regulatory networks of maintaining pluripotency have been described in great detail and, similarly, there is great knowledge on key players that regulate their differentiation. Interestingly, pluripotency has various shades with distinct developmental potential, an observation that coined the term of a ground state of pluripotency. A precise interplay of signalling axes regulates ground state conditions and acts in concert with a combination of key transcription factors. The balance between these transcription factors greatly influences the integrity of the pluripotency network and latest research suggests that minute changes in their expression can strengthen but also collapse the network. Moreover, recent studies reveal different facets of these core factors in balancing a controlled and directed exit from pluripotency. Thereby, subsets of pluripotency-maintaining factors have been shown to adopt new roles during lineage specification and have been globally defined towards neuroectodermal and mesendodermal sets of embryonic stem cell genes. However, detailed underlying insights into how these transcription factors orchestrate cell fate decisions remain largely elusive. Our group and others unravelled complex interactions in the regulation of this controlled exit. Herein, we summarise recent findings and discuss the potential mechanisms involved.
Collapse
|
93
|
Co-culture with mature islet cells augments the differentiation of insulin-producing cells from pluripotent stem cells. Stem Cell Rev Rep 2015; 11:62-74. [PMID: 25173880 DOI: 10.1007/s12015-014-9554-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Islet transplantation has been hampered by the shortage of islet donors available for diabetes therapy. However, pluripotent stem cells (PSCs) can be an alternative source of insulin-producing cells (IPCs) because of their capacity for self-renewal and differentiation. We described a method to efficiently differentiate PSCs into IPCs by co-culturing mature islets with directed-differentiated pancreatic endoderm (PE) cells from mouse and human PSCs. PE cells co-cultured with islet cells or islet cell-derived conditioned medium (CM) showed increased expression levels of β-cell markers; significantly higher levels of proinsulin- and Newport Green (NG)-positive cells, which revealed the characteristics of insulin producing cells; and increased insulin secretion upon glucose stimulation. Co-culturing human PE cells with islet cells was also effective to differentiate PE cells into IPCs. Diabetic nude mice transplanted with co-cultured cells exhibited restored euglycemia, human C-peptide release, and improved glucose tolerance. Immunohistochemistry revealed that insulin+/C-peptide + cells existed in the grafted tissues. These results suggest that mature islet cells can increase the differentiation efficiency of PE cells into mature IPCs via paracrine effects.
Collapse
|
94
|
Kumar N, Richter J, Cutts J, Bush KT, Trujillo C, Nigam SK, Gaasterland T, Brafman D, Willert K. Generation of an expandable intermediate mesoderm restricted progenitor cell line from human pluripotent stem cells. eLife 2015; 4. [PMID: 26554899 PMCID: PMC4631902 DOI: 10.7554/elife.08413] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 10/15/2015] [Indexed: 12/19/2022] Open
Abstract
The field of tissue engineering entered a new era with the development of human pluripotent stem cells (hPSCs), which are capable of unlimited expansion whilst retaining the potential to differentiate into all mature cell populations. However, these cells harbor significant risks, including tumor formation upon transplantation. One way to mitigate this risk is to develop expandable progenitor cell populations with restricted differentiation potential. Here, we used a cellular microarray technology to identify a defined and optimized culture condition that supports the derivation and propagation of a cell population with mesodermal properties. This cell population, referred to as intermediate mesodermal progenitor (IMP) cells, is capable of unlimited expansion, lacks tumor formation potential, and, upon appropriate stimulation, readily acquires properties of a sub-population of kidney cells. Interestingly, IMP cells fail to differentiate into other mesodermally-derived tissues, including blood and heart, suggesting that these cells are restricted to an intermediate mesodermal fate. DOI:http://dx.doi.org/10.7554/eLife.08413.001 The development of ‘human pluripotent stem cells’ has the potential to revolutionize the future of medicine. This is because these cells can both replicate themselves indefinitely (i.e., they can self-renew) and develop into any of the cell types found in the human body (a process that is referred to as differentiation). These abilities mean that the cells could in theory be used to replace any tissues or organs that have been damaged by disease or injury. Unfortunately, transplanting stem cells that are capable of developing into any type of cell comes with the significant risk that these cells will form into a tumor. Once a cell has started to differentiate it can typically only go on to generate a restricted number of cell types. However, these differentiating cells also generally lose their ability to self-renew. Kumar et al. set out to challenge this fundamental property of differentiating cells. A high throughput-screening approach was used to test thousands of combinations of bioactive molecules (i.e., molecules that are known to affect living cells in different ways) to identify some that could promote the self-renewal of cells with a restricted potential to differentiate. Kumar et al. found specific conditions that could cause a population of cells, which they referred to as ‘intermediate mesodermal progenitor cells’ (or IMP cells for short), to self renew. These cells resemble those found in the middle layer of a very early human embryo, which typically go on to develop into only a subset of tissue types in the body—for example, muscle, kidneys and blood vessels, but not brain or lungs. Yet, when Kumar et al. stimulated the self-renewing IMP cells, these cells only differentiated into the cell types that make up the kidney and not any other types of cell. This tight restriction on the differentiation potential of these cells is highly important, because it means that these cells could greatly advance methods to generate kidney cells or even whole kidneys in the laboratory that are suitable for transplantation. DOI:http://dx.doi.org/10.7554/eLife.08413.002
Collapse
Affiliation(s)
- Nathan Kumar
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States
| | - Jenna Richter
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States
| | - Josh Cutts
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, United States
| | - Kevin T Bush
- Department of Pediatrics, University of California, San Diego, San Diego, United States
| | - Cleber Trujillo
- Department of Pediatrics, University of California, San Diego, San Diego, United States
| | - Sanjay K Nigam
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States.,Department of Pediatrics, University of California, San Diego, San Diego, United States
| | - Terry Gaasterland
- Scripps Institution of Oceanography, Scripps Genome Center, University of California, San Diego, San Diego, United States
| | - David Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, United States
| | - Karl Willert
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States
| |
Collapse
|
95
|
Ignatius Irudayam J, Contreras D, Spurka L, Ren S, Kanagavel V, Ramaiah A, Annamalai A, French SW, Klein AS, Funari V, Arumugaswami V. Profile of Inflammation-associated genes during Hepatic Differentiation of Human Pluripotent Stem Cells. Data Brief 2015; 5:871-8. [PMID: 26702414 PMCID: PMC4669432 DOI: 10.1016/j.dib.2015.10.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 10/14/2015] [Accepted: 10/22/2015] [Indexed: 12/12/2022] Open
Abstract
Expression of genes associated with inflammation was analyzed during differentiation of human pluripotent stem cells (PSCs) to hepatic cells. Messenger RNA transcript profiles of differentiated endoderm (day 5), hepatoblast (day 15) and hepatocyte-like cells (day 21) were obtained by RNA sequencing analysis. When compared to endoderm cells an immature cell type, the hepatic cells (days 15 and 21) had significantly higher expression of acute phase protein genes including complement factors, coagulation factors, serum amyloid A and serpins. Furthermore, hepatic phase of cells expressed proinflammatory cytokines IL18 and IL32 as well as cytokine receptors IL18R1, IL1R1, IL1RAP, IL2RG, IL6R, IL6ST and IL10RB. These cells also produced CCL14, CCL15, and CXCL- 1, 2, 3, 16 and 17 chemokines. Endoderm cells had higher levels of chemokine receptors, CXCR4 and CXCR7, than that of hepatic cells. Sirtuin family of genes involved in aging, inflammation and metabolism were differentially regulated in endoderm and hepatic phase cells. Ligands and receptors of the tumor necrosis factor (TNF) family as well as downstream signaling factors TRAF2, TRAF4, FADD, NFKB1 and NFKBIB were differentially expressed during hepatic differentiation.
Collapse
Affiliation(s)
- Joseph Ignatius Irudayam
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Deisy Contreras
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Lindsay Spurka
- Cedars-Sinai Genomics Core, Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Songyang Ren
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Vidhya Kanagavel
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Arunachalam Ramaiah
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, Karnataka 560012, India ; Hindustan Genomics Institute, SVA Medical Center, Kadayam, Tamil Nadu 627415, India
| | - Alagappan Annamalai
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Samuel W French
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Andrew S Klein
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA ; Department of Surgery, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Vincent Funari
- Cedars-Sinai Genomics Core, Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Vaithilingaraja Arumugaswami
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA ; Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA ; Department of Surgery, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
96
|
Busser BW, Lin Y, Yang Y, Zhu J, Chen G, Michelson AM. An Orthologous Epigenetic Gene Expression Signature Derived from Differentiating Embryonic Stem Cells Identifies Regulators of Cardiogenesis. PLoS One 2015; 10:e0141066. [PMID: 26485529 PMCID: PMC4617299 DOI: 10.1371/journal.pone.0141066] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/05/2015] [Indexed: 01/18/2023] Open
Abstract
Here we used predictive gene expression signatures within a multi-species framework to identify the genes that underlie cardiac cell fate decisions in differentiating embryonic stem cells. We show that the overlapping orthologous mouse and human genes are the most accurate candidate cardiogenic genes as these genes identified the most conserved developmental pathways that characterize the cardiac lineage. An RNAi-based screen of the candidate genes in Drosophila uncovered numerous novel cardiogenic genes. shRNA knockdown combined with transcriptome profiling of the newly-identified transcription factors zinc finger protein 503 and zinc finger E-box binding homeobox 2 and the well-known cardiac regulatory factor NK2 homeobox 5 revealed that zinc finger E-box binding homeobox 2 activates terminal differentiation genes required for cardiomyocyte structure and function whereas zinc finger protein 503 and NK2 homeobox 5 are required for specification of the cardiac lineage. We further demonstrated that an essential role of NK2 homeobox 5 and zinc finger protein 503 in specification of the cardiac lineage is the repression of gene expression programs characteristic of alternative cell fates. Collectively, these results show that orthologous gene expression signatures can be used to identify conserved cardiogenic pathways.
Collapse
Affiliation(s)
- Brian W. Busser
- Systems Biology Center, Division of Intramural Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, United States of America
- * E-mail: (AMM); (BWB)
| | - Yongshun Lin
- Center for Molecular Medicine, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, United States of America
| | - Yanqin Yang
- Systems Biology Center, Division of Intramural Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, United States of America
| | - Jun Zhu
- Systems Biology Center, Division of Intramural Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, United States of America
| | - Guokai Chen
- Center for Molecular Medicine, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, United States of America
| | - Alan M. Michelson
- Systems Biology Center, Division of Intramural Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, United States of America
- * E-mail: (AMM); (BWB)
| |
Collapse
|
97
|
Rostovskaya M, Bredenkamp N, Smith A. Towards consistent generation of pancreatic lineage progenitors from human pluripotent stem cells. Philos Trans R Soc Lond B Biol Sci 2015; 370:20140365. [PMID: 26416676 PMCID: PMC4633994 DOI: 10.1098/rstb.2014.0365] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2015] [Indexed: 12/12/2022] Open
Abstract
Human pluripotent stem cells can in principle be used as a source of any differentiated cell type for disease modelling, drug screening, toxicology testing or cell replacement therapy. Type I diabetes is considered a major target for stem cell applications due to the shortage of primary human beta cells. Several protocols have been reported for generating pancreatic progenitors by in vitro differentiation of human pluripotent stem cells. Here we first assessed one of these protocols on a panel of pluripotent stem cell lines for capacity to engender glucose sensitive insulin-producing cells after engraftment in immunocompromised mice. We observed variable outcomes with only one cell line showing a low level of glucose response. We, therefore, undertook a systematic comparison of different methods for inducing definitive endoderm and subsequently pancreatic differentiation. Of several protocols tested, we identified a combined approach that robustly generated pancreatic progenitors in vitro from both embryo-derived and induced pluripotent stem cells. These findings suggest that, although there are intrinsic differences in lineage specification propensity between pluripotent stem cell lines, optimal differentiation procedures may consistently direct a substantial fraction of cells into pancreatic specification.
Collapse
Affiliation(s)
- Maria Rostovskaya
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Nicholas Bredenkamp
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Austin Smith
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| |
Collapse
|
98
|
Oh K, Shon SY, Seo MW, Lee HM, Oh JE, Choi EY, Lee DS, Park KS. Murine Sca1(+)Lin(-) bone marrow contains an endodermal precursor population that differentiates into hepatocytes. Exp Mol Med 2015; 47:e187. [PMID: 26427852 PMCID: PMC4673473 DOI: 10.1038/emm.2015.64] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/26/2015] [Accepted: 06/01/2015] [Indexed: 12/12/2022] Open
Abstract
The direct differentiation of hepatocytes from bone marrow cells remains controversial. Several mechanisms, including transdifferentiation and cell fusion, have been proposed for this phenomenon, although direct visualization of the process and the underlying mechanisms have not been reported. In this study, we established an efficient in vitro culture method for differentiation of functioning hepatocytes from murine lineage-negative bone marrow cells. These cells reduced liver damage and incorporated into hepatic parenchyma in two independent hepatic injury models. Our simple and efficient in vitro protocol for endodermal precursor cell survival and expansion enabled us to identify these cells as existing in Sca1+ subpopulations of lineage-negative bone marrow cells. The endodermal precursor cells followed a sequential developmental pathway that included endodermal cells and hepatocyte precursor cells, which indicates that lineage-negative bone marrow cells contain more diverse multipotent stem cells than considered previously. The presence of equivalent endodermal precursor populations in human bone marrow would facilitate the development of these cells into an effective treatment modality for chronic liver diseases.
Collapse
Affiliation(s)
- Keunhee Oh
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.,Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Suh Youn Shon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Myung Won Seo
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.,Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hak Mo Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Ju-Eun Oh
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Dong-Sup Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.,Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Kyong Soo Park
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| |
Collapse
|
99
|
Ying L, Mills JA, French DL, Gadue P. OCT4 Coordinates with WNT Signaling to Pre-pattern Chromatin at the SOX17 Locus during Human ES Cell Differentiation into Definitive Endoderm. Stem Cell Reports 2015; 5:490-8. [PMID: 26411902 PMCID: PMC4624996 DOI: 10.1016/j.stemcr.2015.08.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 08/25/2015] [Accepted: 08/26/2015] [Indexed: 12/02/2022] Open
Abstract
We demonstrate that the pluripotency gene OCT4 has a role in regulating differentiation via Wnt signaling. OCT4 expression levels in human embryonic stem cells increases transiently during the first 24 hr of in vitro differentiation, with OCT4 occupancy increasing at endoderm regulators such as SOX17 and FOXA2. This increased occupancy correlates with loss of the PRC2 complex and the inhibitory histone mark H3K27me3. Knockdown of OCT4 during differentiation inhibits mesendoderm formation and removal of the H3K27me3 mark from the SOX17 promoter, suggesting that OCT4 acts to induce removal of the PRC2 complex. Furthermore, OCT4 and β-catenin can be co-immunoprecipitated upon differentiation, and Wnt stimulation is required for the enhanced OCT4 occupancy and loss of the PRC2 complex from the SOX17 promoter. In conclusion, our study reveals that OCT4, a master regulator of pluripotency, may also collaborate with Wnt signaling to drive endoderm induction by pre-patterning epigenetic markers on endodermal promoters. OCT4 occupancy increases at endoderm genes early in ES cell differentiation The PRC2 complex is lost from OCT4 sites on endoderm genes during differentiation OCT4 associates with β-catenin during ES cell differentiation OCT4 and Wnt are both required for mesendoderm induction
Collapse
Affiliation(s)
- Lei Ying
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jason A Mills
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Deborah L French
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paul Gadue
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
100
|
Two Effective Routes for Removing Lineage Restriction Roadblocks: From Somatic Cells to Hepatocytes. Int J Mol Sci 2015; 16:20873-95. [PMID: 26340624 PMCID: PMC4613233 DOI: 10.3390/ijms160920873] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 08/24/2015] [Accepted: 08/24/2015] [Indexed: 12/31/2022] Open
Abstract
The conversion of somatic cells to hepatocytes has fundamentally re-shaped traditional concepts regarding the limited resources for hepatocyte therapy. With the various induced pluripotent stem cell (iPSC) generation routes, most somatic cells can be effectively directed to functional stem cells, and this strategy will supply enough pluripotent material to generate promising functional hepatocytes. However, the major challenges and potential applications of reprogrammed hepatocytes remain under investigation. In this review, we provide a summary of two effective routes including direct reprogramming and indirect reprogramming from somatic cells to hepatocytes and the general potential applications of the resulting hepatocytes. Through these approaches, we are striving toward the goal of achieving a robust, mature source of clinically relevant lineages.
Collapse
|