1
|
Aydin O, Passaro AP, Raman R, Spellicy SE, Weinberg RP, Kamm RD, Sample M, Truskey GA, Zartman J, Dar RD, Palacios S, Wang J, Tordoff J, Montserrat N, Bashir R, Saif MTA, Weiss R. Principles for the design of multicellular engineered living systems. APL Bioeng 2022; 6:010903. [PMID: 35274072 PMCID: PMC8893975 DOI: 10.1063/5.0076635] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/02/2022] [Indexed: 12/14/2022] Open
Abstract
Remarkable progress in bioengineering over the past two decades has enabled the formulation of fundamental design principles for a variety of medical and non-medical applications. These advancements have laid the foundation for building multicellular engineered living systems (M-CELS) from biological parts, forming functional modules integrated into living machines. These cognizant design principles for living systems encompass novel genetic circuit manipulation, self-assembly, cell-cell/matrix communication, and artificial tissues/organs enabled through systems biology, bioinformatics, computational biology, genetic engineering, and microfluidics. Here, we introduce design principles and a blueprint for forward production of robust and standardized M-CELS, which may undergo variable reiterations through the classic design-build-test-debug cycle. This Review provides practical and theoretical frameworks to forward-design, control, and optimize novel M-CELS. Potential applications include biopharmaceuticals, bioreactor factories, biofuels, environmental bioremediation, cellular computing, biohybrid digital technology, and experimental investigations into mechanisms of multicellular organisms normally hidden inside the "black box" of living cells.
Collapse
Affiliation(s)
| | - Austin P. Passaro
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia 30602, USA
| | - Ritu Raman
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | - Robert P. Weinberg
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston, Massachusetts 02115, USA
| | | | - Matthew Sample
- Center for Ethics and Law in the Life Sciences, Leibniz Universität Hannover, 30167 Hannover, Germany
| | - George A. Truskey
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Jeremiah Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Roy D. Dar
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Sebastian Palacios
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Jason Wang
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Jesse Tordoff
- Computational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Nuria Montserrat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | | | - M. Taher A. Saif
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Ron Weiss
- Author to whom correspondence should be addressed:
| |
Collapse
|
2
|
Lewis-Israeli YR, Wasserman AH, Gabalski MA, Volmert BD, Ming Y, Ball KA, Yang W, Zou J, Ni G, Pajares N, Chatzistavrou X, Li W, Zhou C, Aguirre A. Self-assembling human heart organoids for the modeling of cardiac development and congenital heart disease. Nat Commun 2021; 12:5142. [PMID: 34446706 PMCID: PMC8390749 DOI: 10.1038/s41467-021-25329-5] [Citation(s) in RCA: 255] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023] Open
Abstract
Congenital heart defects constitute the most common human birth defect, however understanding of how these disorders originate is limited by our ability to model the human heart accurately in vitro. Here we report a method to generate developmentally relevant human heart organoids by self-assembly using human pluripotent stem cells. Our procedure is fully defined, efficient, reproducible, and compatible with high-content approaches. Organoids are generated through a three-step Wnt signaling modulation strategy using chemical inhibitors and growth factors. Heart organoids are comparable to age-matched human fetal cardiac tissues at the transcriptomic, structural, and cellular level. They develop sophisticated internal chambers with well-organized multi-lineage cardiac cell types, recapitulate heart field formation and atrioventricular specification, develop a complex vasculature, and exhibit robust functional activity. We also show that our organoid platform can recreate complex metabolic disorders associated with congenital heart defects, as demonstrated by an in vitro model of pregestational diabetes-induced congenital heart defects.
Collapse
Affiliation(s)
- Yonatan R Lewis-Israeli
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Aaron H Wasserman
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Mitchell A Gabalski
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Brett D Volmert
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Yixuan Ming
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, USA
| | - Kristen A Ball
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Weiyang Yang
- Division of Biomedical Devices, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Electrical and Computer Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Jinyun Zou
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, USA
| | - Guangming Ni
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, USA
| | - Natalia Pajares
- Department of Chemical Engineering and Material Science, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Xanthippi Chatzistavrou
- Department of Chemical Engineering and Material Science, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Wen Li
- Division of Biomedical Devices, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Electrical and Computer Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Chao Zhou
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, USA
| | - Aitor Aguirre
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA.
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
3
|
Bruveris FF, Ng ES, Stanley EG, Elefanty AG. VEGF, FGF2, and BMP4 regulate transitions of mesoderm to endothelium and blood cells in a human model of yolk sac hematopoiesis. Exp Hematol 2021; 103:30-39.e2. [PMID: 34437953 DOI: 10.1016/j.exphem.2021.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022]
Abstract
Exogenous growth factors play an important role in mediating hematopoietic differentiation of human pluripotent stem cells. We explored the role of different factors in early human blood cell production using blast colony formation in methylcellulose as a surrogate assay for yolk sac hematopoiesis. A reporter cell line that read out endothelial (SOX17+) and hematopoietic (RUNX1C+) progenitors facilitated the identification of basic fibroblast growth and vascular endothelial growth factor as critical signals for the progression of mesoderm into endothelium. Bone morphogenetic protein 4 was needed for the subsequent generation of blood from hemogenic endothelium, and this was antagonized by Activin A or high concentrations of the WNT agonist CHIR-99021. Manipulations of the Hedgehog pathway or inhibition of Notch signaling reduced blast colony frequency but did not perturb cell differentiation. These data help to define distinct roles for prerequisite growth factors that commit mesoderm to hemogenic endothelium and subsequently allocate cells to blood lineages.
Collapse
Affiliation(s)
- Freya F Bruveris
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth S Ng
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria, Australia
| | - Edouard G Stanley
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Andrew G Elefanty
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
4
|
Stavish D, Böiers C, Price C, Frith TJR, Halliwell J, Saldaña-Guerrero I, Wray J, Brown J, Carr J, James C, Barbaric I, Andrews PW, Enver T. Generation and trapping of a mesoderm biased state of human pluripotency. Nat Commun 2020; 11:4989. [PMID: 33020476 PMCID: PMC7536399 DOI: 10.1038/s41467-020-18727-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 09/10/2020] [Indexed: 12/22/2022] Open
Abstract
We postulate that exit from pluripotency involves intermediates that retain pluripotency while simultaneously exhibiting lineage-bias. Using a MIXL1 reporter, we explore mesoderm lineage-bias within the human pluripotent stem cell compartment. We identify a substate, which at the single cell level coexpresses pluripotent and mesodermal gene expression programmes. Functionally these cells initiate stem cell cultures and exhibit mesodermal bias in differentiation assays. By promoting mesodermal identity through manipulation of WNT signalling while preventing exit from pluripotency using lysophosphatidic acid, we 'trap' and maintain cells in a lineage-biased stem cell state through multiple passages. These cells correspond to a normal state on the differentiation trajectory, the plasticity of which is evidenced by their reacquisition of an unbiased state upon removal of differentiation cues. The use of 'cross-antagonistic' signalling to trap pluripotent stem cell intermediates with different lineage-bias may have general applicability in the efficient production of cells for regenerative medicine.
Collapse
Affiliation(s)
- Dylan Stavish
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
| | - Charlotta Böiers
- Stem Cell Laboratory, Department of Cancer Biology, University College London Cancer Institute, 72 Huntley St, London, WC1E 6AG, UK
| | - Christopher Price
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Thomas J R Frith
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Jason Halliwell
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Ingrid Saldaña-Guerrero
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Jason Wray
- Stem Cell Laboratory, Department of Cancer Biology, University College London Cancer Institute, 72 Huntley St, London, WC1E 6AG, UK
| | - John Brown
- Stem Cell Laboratory, Department of Cancer Biology, University College London Cancer Institute, 72 Huntley St, London, WC1E 6AG, UK
| | - Jonathon Carr
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Chela James
- Stem Cell Laboratory, Department of Cancer Biology, University College London Cancer Institute, 72 Huntley St, London, WC1E 6AG, UK
| | - Ivana Barbaric
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Peter W Andrews
- The Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
| | - Tariq Enver
- Stem Cell Laboratory, Department of Cancer Biology, University College London Cancer Institute, 72 Huntley St, London, WC1E 6AG, UK
| |
Collapse
|
5
|
Gaspari E, Franke A, Robles-Diaz D, Zweigerdt R, Roeder I, Zerjatke T, Kempf H. Paracrine mechanisms in early differentiation of human pluripotent stem cells: Insights from a mathematical model. Stem Cell Res 2018; 32:1-7. [PMID: 30145492 DOI: 10.1016/j.scr.2018.07.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/13/2018] [Accepted: 07/24/2018] [Indexed: 02/01/2023] Open
Abstract
With their capability to self-renew and differentiate into derivatives of all three germ layers, human pluripotent stem cells (hPSCs) offer a unique model to study aspects of human development in vitro. Directed differentiation towards mesendodermal lineages is a complex process, involving transition through a primitive streak (PS)-like stage. We have recently shown PS-like patterning from hPSCs into definitive endoderm, cardiac as well as presomitic mesoderm by only modulating the bulk cell density and the concentration of the GSK3 inhibitor CHIR99021, a potent activator of the WNT pathway. The patterning process is modulated by a complex paracrine network, whose identity and mechanistic consequences are poorly understood. To study the underlying dynamics, we here applied mathematical modeling based on ordinary differential equations. We compared time-course data of early hPSC differentiation to increasingly complex model structures with incremental numbers of paracrine factors. Model simulations suggest at least three paracrine factors being required to recapitulate the experimentally observed differentiation kinetics. Feedback mechanisms from both undifferentiated and differentiated cells turned out to be crucial. Evidence from double knock-down experiments and secreted protein enrichment allowed us to hypothesize on the identity of two of the three predicted factors. From a practical perspective, the mathematical model predicts optimal settings for directing lineage-specific differentiation. This opens new avenues for rational stem cell bioprocessing in more advanced culture systems, e.g. in perfusion-fed bioreactors enabling cell therapies.
Collapse
Affiliation(s)
- Erika Gaspari
- Institute for Medical Informatics and Biometry, Carl Gustav Carus Faculty of Medicine, TU Dresden, Dresden, Germany; Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Wageningen, the Netherlands
| | - Annika Franke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Cluster of Excellence, Hannover Medical School, Germany
| | - Diana Robles-Diaz
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Cluster of Excellence, Hannover Medical School, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Cluster of Excellence, Hannover Medical School, Germany
| | - Ingo Roeder
- Institute for Medical Informatics and Biometry, Carl Gustav Carus Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Thomas Zerjatke
- Institute for Medical Informatics and Biometry, Carl Gustav Carus Faculty of Medicine, TU Dresden, Dresden, Germany.
| | - Henning Kempf
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Cluster of Excellence, Hannover Medical School, Germany.
| |
Collapse
|
6
|
Rico-Varela J, Ho D, Wan LQ. In Vitro Microscale Models for Embryogenesis. ADVANCED BIOSYSTEMS 2018; 2:1700235. [PMID: 30533517 PMCID: PMC6286056 DOI: 10.1002/adbi.201700235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Indexed: 12/15/2022]
Abstract
Embryogenesis is a highly regulated developmental process requiring complex mechanical and biochemical microenvironments to give rise to a fully developed and functional embryo. Significant efforts have been taken to recapitulate specific features of embryogenesis by presenting the cells with developmentally relevant signals. The outcomes, however, are limited partly due to the complexity of this biological process. Microtechnologies such as micropatterned and microfluidic systems, along with new emerging embryonic stem cell-based models, could potentially serve as powerful tools to study embryogenesis. The aim of this article is to review major studies involving the culturing of pluripotent stem cells using different geometrical patterns, microfluidic platforms, and embryo/embryoid body-on-a-chip modalities. Indeed, new research opportunities have emerged for establishing in vitro culture for studying human embryogenesis and for high-throughput pharmacological testing platforms and disease models to prevent defects in early stages of human development.
Collapse
Affiliation(s)
- Jennifer Rico-Varela
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
| | - Dominic Ho
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
| | - Leo Q. Wan
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
- Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
- Center for Modeling, Simulation and Imaging in Medicine, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
| |
Collapse
|
7
|
Russell RP, Fu Y, Liu Y, Maye P. Inverse agonism of retinoic acid receptors directs epiblast cells into the paraxial mesoderm lineage. Stem Cell Res 2018; 30:85-95. [PMID: 29807258 PMCID: PMC6083448 DOI: 10.1016/j.scr.2018.05.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/04/2018] [Accepted: 05/21/2018] [Indexed: 01/16/2023] Open
Abstract
We have investigated the differentiation of paraxial mesoderm from mouse embryonic stem cells utilizing a Tbx6-EYFP/Brachyury (T)-Cherry dual reporter system. Differentiation from the mouse ESC state directly into mesoderm via Wnt pathway activation was low, but augmented by treatment with AGN193109, a pan-retinoic acid receptor inverse agonist. After five days of differentiation, T+ cells increased from 12.2% to 18.8%, Tbx6+ cells increased from 5.8% to 12.7%, and T+/Tbx6+ cells increased from 2.4% to 14.1%. The synergism of AGN193109 with Wnt3a/CHIR99021 was further substantiated by the increased expression of paraxial mesoderm gene markers Tbx6, Msgn1, Meox1, and Hoxb1. Separate to inverse agonist treatment, when mouse ESCs were indirectly differentiated into mesoderm via a transient epiblast step the efficiency of paraxial mesoderm formation markedly increased. Tbx6+ cells represented 65-75% of the total cell population after just 3 days of differentiation and the expression of paraxial mesoderm marker genes Tbx6 and Msgn increased over 100-fold and 300-fold, respectively. Further evaluation of AGN193109 treatment on the indirect differentiation protocol suggested that RARs have two distinct roles. First, AGN193109 treatment at the epiblast step and mesoderm step promoted paraxial mesoderm formation over other mesoderm and endoderm lineage types. Second, continued treatment during mesoderm formation revealed its ability to repress the maturation of presomitic mesoderm into somitic paraxial mesoderm. Thus, the continuous treatment of AGN193109 during epiblast and mesoderm differentiation steps yielded a culture where ~90% of the cells were Tbx6+. The surprisingly early effect of inverse agonist treatment at the epiblast step of differentiation led us to further examine the effect of AGN193109 treatment during an extended epiblast differentiation protocol. Interestingly, while inverse agonist treatment had no impact on the conversion of ESCs into epiblast cells based on the expression of Rex1, Fgf5, and pluripotency marker genes Oct4, Nanog, and Sox2, after three days of differentiation in the presence of AGN193109 caudal epiblast and early paraxial mesoderm marker genes, T, Cyp26a1, Fgf8, Tbx6 and Msgn were all highly up-regulated. Collectively, our studies reveal an earlier than appreciated role for RARs in epiblast cells and the modulation of their function via inverse agonist treatment can promote their differentiation into the paraxial mesoderm lineage.
Collapse
Affiliation(s)
- Ryan P Russell
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, United States
| | - Yu Fu
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, United States
| | - Yaling Liu
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, United States
| | - Peter Maye
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, United States.
| |
Collapse
|
8
|
WNT9A Is a Conserved Regulator of Hematopoietic Stem and Progenitor Cell Development. Genes (Basel) 2018; 9:genes9020066. [PMID: 29382179 PMCID: PMC5852562 DOI: 10.3390/genes9020066] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/10/2018] [Accepted: 01/23/2018] [Indexed: 02/08/2023] Open
Abstract
Hematopoietic stem cells (HSCs) differentiate into all cell types of the blood and can be used therapeutically to treat hematopoietic cancers and disorders. Despite decades of research, it is not yet possible to derive therapy-grade HSCs from pluripotent precursors. Analysis of HSC development in model organisms has identified some of the molecular cues that are necessary to instruct hematopoiesis in vivo, including Wnt9A, which is required during an early time window in zebrafish development. Although bona fide HSCs cannot be derived in vitro, it is possible to model human hematopoietic progenitor development by differentiating human pluripotent stem cells to hematopoietic cells. Herein, we modulate WNT9A expression during the in vitro differentiation of human embryonic stem cells to hematopoietic progenitor cells and demonstrate that WNT9A also regulates human hematopoietic progenitor cell development in vitro. Overexpression of WNT9A only impacts differentiation to CD34+/CD45+ cells during early time windows and does so in a dose-dependent manner. The cells that receive the Wnt signal—not the cells that secrete WNT9A—differentiate most efficiently to hematopoietic progenitors; this mimics the paracrine action of Wnt9a during in vivo hematopoiesis. Taken together, these data indicate that WNT9A is a conserved regulator of zebrafish and human hematopoietic development.
Collapse
|
9
|
Tachikawa S, Nishimura T, Nakauchi H, Ohnuma K. Thalidomide induces apoptosis in undifferentiated human induced pluripotent stem cells. In Vitro Cell Dev Biol Anim 2017; 53:841-851. [PMID: 28849348 DOI: 10.1007/s11626-017-0192-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/28/2017] [Indexed: 10/19/2022]
Abstract
Thalidomide, which was formerly available commercially to control the symptoms of morning sickness, is a strong teratogen that causes fetal abnormalities. However, the mechanism of thalidomide teratogenicity is not fully understood; thalidomide toxicity is not apparent in rodents, and the use of human embryos is ethically and technically untenable. In this study, we designed an experimental system featuring human-induced pluripotent stem cells (hiPSCs) to investigate the effects of thalidomide. These cells exhibit the same characteristics as those of epiblasts originating from implanted fertilized ova, which give rise to the fetus. Therefore, theoretically, thalidomide exposure during hiPSC differentiation is equivalent to that in the human fetus. We examined the effects of thalidomide on undifferentiated hiPSCs and early-differentiated hiPSCs cultured in media containing bone morphogenetic protein-4, which correspond, respectively, to epiblast (future fetus) and trophoblast (future extra-embryonic tissue). We found that only the number of undifferentiated cells was reduced. In undifferentiated cells, application of thalidomide increased the number of apoptotic and dead cells at day 2 but not day 4. Application of thalidomide did not affect the cell cycle. Furthermore, immunostaining and flow cytometric analysis revealed that thalidomide exposure had no effect on the expression of specific markers of undifferentiated and early trophectodermal differentiated cells. These results suggest that the effect of thalidomide was successfully detected in our experimental system and that thalidomide eliminated a subpopulation of undifferentiated hiPSCs. This study may help to elucidate the mechanisms underlying thalidomide teratogenicity and reveal potential strategies for safely prescribing this drug to pregnant women.
Collapse
Affiliation(s)
- Saoko Tachikawa
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan
| | - Toshinobu Nishimura
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Kiyoshi Ohnuma
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan. .,Department of Science of Technology Innovation, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan.
| |
Collapse
|
10
|
Peters EB. Endothelial Progenitor Cells for the Vascularization of Engineered Tissues. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:1-24. [PMID: 28548628 DOI: 10.1089/ten.teb.2017.0127] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Self-assembled microvasculature from cocultures of endothelial cells (ECs) and stromal cells has significantly advanced efforts to vascularize engineered tissues by enhancing perfusion rates in vivo and producing investigative platforms for microvascular morphogenesis in vitro. However, to clinically translate prevascularized constructs, the issue of EC source must be resolved. Endothelial progenitor cells (EPCs) can be noninvasively supplied from the recipient through adult peripheral and umbilical cord blood, as well as derived from induced pluripotent stem cells, alleviating antigenicity issues. EPCs can also differentiate into all tissue endothelium, and have demonstrated potential for therapeutic vascularization. Yet, EPCs are not the standard EC choice to vascularize tissue constructs in vitro. Possible reasons include unresolved issues with EPC identity and characterization, as well as uncertainty in the selection of coculture, scaffold, and culture media combinations that promote EPC microvessel formation. This review addresses these issues through a summary of EPC vascular biology and the effects of tissue engineering design parameters upon EPC microvessel formation. Also included are perspectives to integrate EPCs with emerging technologies to produce functional, organotypic vascularized tissues.
Collapse
Affiliation(s)
- Erica B Peters
- Department of Chemical and Biological Engineering, University of Colorado , Boulder, Colorado
| |
Collapse
|
11
|
Tesarova L, Simara P, Stejskal S, Koutna I. Hematopoietic Developmental Potential of Human Pluripotent Stem Cell Lines Is Accompanied by the Morphology of Embryoid Bodies and the Expression of Endodermal and Hematopoietic Markers. Cell Reprogram 2017. [PMID: 28632430 DOI: 10.1089/cell.2016.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The potential clinical applications of hematopoietic stem cells (HSCs) derived from human pluripotent stem cells (hPSCs) are limited by the difficulty of recapitulating embryoid hematopoiesis and by the unknown differentiation potential of hPSC lines. To evaluate their hematopoietic developmental potential, available hPSC lines were differentiated by an embryoid body (EB) suspension culture in serum-free medium supplemented with three different cytokine mixes (CMs). The hPSC differentiation status was investigated by the flow cytometry expression profiles of cell surface molecules, and the gene expression of pluripotency and differentiation markers over time was evaluated by real-time reverse transcription polymerase chain reaction (qRT-PCR). hPSC lines differed in several aspects of the differentiation process, including the absolute yield of hematopoietic progenitors, the proportion of hematopoietic progenitor populations, and the effect of various CMs. The ability to generate hematopoietic progenitors was then associated with the morphology of the developing EBs, the expression of the endodermal markers AFP and SOX17, and the hematopoietic transcription factor RUNX1. These findings deepen the knowledge about the hematopoietic propensity of hPSCs and identify its variability as an aspect that must be taken into account before the usage of hPSC-derived HSCs in downstream applications.
Collapse
Affiliation(s)
- Lenka Tesarova
- 1 Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University , Brno, Czech Republic .,2 International Clinical Research Center, St. Anne's University Hospital Brno , Brno, Czech Republic
| | - Pavel Simara
- 1 Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University , Brno, Czech Republic .,2 International Clinical Research Center, St. Anne's University Hospital Brno , Brno, Czech Republic
| | - Stanislav Stejskal
- 1 Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University , Brno, Czech Republic
| | - Irena Koutna
- 1 Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University , Brno, Czech Republic .,2 International Clinical Research Center, St. Anne's University Hospital Brno , Brno, Czech Republic
| |
Collapse
|
12
|
Chen ACH, Lee YL, Fong SW, Wong CCY, Ng EHY, Yeung WSB. Hyperglycemia impedes definitive endoderm differentiation of human embryonic stem cells by modulating histone methylation patterns. Cell Tissue Res 2017; 368:563-578. [PMID: 28283910 DOI: 10.1007/s00441-017-2583-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 01/27/2017] [Indexed: 12/25/2022]
Abstract
Exposure to maternal diabetes during fetal growth is a risk factor for the development of type II diabetes (T2D) in later life. Discovery of the mechanisms involved in this association should provide valuable background for therapeutic treatments. Early embryogenesis involves epigenetic changes including histone modifications. The bivalent histone methylation marks H3K4me3 and H3K27me3 are important for regulating key developmental genes during early fetal pancreas specification. We hypothesized that maternal hyperglycemia disrupted early pancreas development through changes in histone bivalency. A human embryonic stem cell line (VAL3) was used as the cell model for studying the effects of hyperglycemia upon differentiation into definitive endoderm (DE), an early stage of the pancreatic lineage. Hyperglycemic conditions significantly down-regulated the expression levels of DE markers SOX17, FOXA2, CXCR4 and EOMES during differentiation. This was associated with retention of the repressive histone methylation mark H3K27me3 on their promoters under hyperglycemic conditions. The disruption of histone methylation patterns was observed as early as the mesendoderm stage, with Wnt/β-catenin signaling being suppressed during hyperglycemia. Treatment with Wnt/β-catenin signaling activator CHIR-99021 restored the expression levels and chromatin methylation status of DE markers, even in a hyperglycemic environment. The disruption of DE development was also found in mouse embryos at day 7.5 post coitum from diabetic mothers. Furthermore, disruption of DE differentiation in VAL3 cells led to subsequent impairment in pancreatic progenitor formation. Thus, early exposure to hyperglycemic conditions hinders DE development with a possible relationship to the later impairment of pancreas specification.
Collapse
Affiliation(s)
- A C H Chen
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China
| | - Y L Lee
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China.
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, The University of Hong Kong, Shenzhen, People's Republic of China.
- Center for Reproduction, Development and Growth, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China.
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Room 747, 21 Sassoon Road, Hong Kong, SAR, People's Republic of China.
| | - S W Fong
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China
| | - C C Y Wong
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China
| | - E H Y Ng
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, The University of Hong Kong, Shenzhen, People's Republic of China
- Center for Reproduction, Development and Growth, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China
| | - W S B Yeung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, The University of Hong Kong, Shenzhen, People's Republic of China
- Center for Reproduction, Development and Growth, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China
| |
Collapse
|
13
|
Links between DNA Replication, Stem Cells and Cancer. Genes (Basel) 2017; 8:genes8020045. [PMID: 28125050 PMCID: PMC5333035 DOI: 10.3390/genes8020045] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 01/02/2017] [Accepted: 01/12/2017] [Indexed: 12/31/2022] Open
Abstract
Cancers can be categorized into two groups: those whose frequency increases with age, and those resulting from errors during mammalian development. The first group is linked to DNA replication through the accumulation of genetic mutations that occur during proliferation of developmentally acquired stem cells that give rise to and maintain tissues and organs. These mutations, which result from DNA replication errors as well as environmental insults, fall into two categories; cancer driver mutations that initiate carcinogenesis and genome destabilizing mutations that promote aneuploidy through excess genome duplication and chromatid missegregation. Increased genome instability results in accelerated clonal evolution leading to the appearance of more aggressive clones with increased drug resistance. The second group of cancers, termed germ cell neoplasia, results from the mislocation of pluripotent stem cells during early development. During normal development, pluripotent stem cells that originate in early embryos give rise to all of the cell lineages in the embryo and adult, but when they mislocate to ectopic sites, they produce tumors. Remarkably, pluripotent stem cells, like many cancer cells, depend on the Geminin protein to prevent excess DNA replication from triggering DNA damage-dependent apoptosis. This link between the control of DNA replication during early development and germ cell neoplasia reveals Geminin as a potential chemotherapeutic target in the eradication of cancer progenitor cells.
Collapse
|
14
|
Activin A Modulates CRIPTO-1/HNF4 α+ Cells to Guide Cardiac Differentiation from Human Embryonic Stem Cells. Stem Cells Int 2017; 2017:4651238. [PMID: 28163723 PMCID: PMC5253508 DOI: 10.1155/2017/4651238] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/29/2016] [Accepted: 12/01/2016] [Indexed: 01/02/2023] Open
Abstract
The use of human pluripotent stem cells in basic and translational cardiac research requires efficient differentiation protocols towards cardiomyocytes. In vitro differentiation yields heterogeneous populations of ventricular-, atrial-, and nodal-like cells hindering their potential applications in regenerative therapies. We described the effect of the growth factor Activin A during early human embryonic stem cell fate determination in cardiac differentiation. Addition of high levels of Activin A during embryoid body cardiac differentiation augmented the generation of endoderm derivatives, which in turn promoted cardiomyocyte differentiation. Moreover, a dose-dependent increase in the coreceptor expression of the TGF-β superfamily member CRIPTO-1 was observed in response to Activin A. We hypothesized that interactions between cells derived from meso- and endodermal lineages in embryoid bodies contributed to improved cell maturation in early stages of cardiac differentiation, improving the beating frequency and the percentage of contracting embryoid bodies. Activin A did not seem to affect the properties of cardiomyocytes at later stages of differentiation, measuring action potentials, and intracellular Ca2+ dynamics. These findings are relevant for improving our understanding on human heart development, and the proposed protocol could be further explored to obtain cardiomyocytes with functional phenotypes, similar to those observed in adult cardiac myocytes.
Collapse
|
15
|
Kim N, Minami N, Yamada M, Imai H. Immobilized pH in culture reveals an optimal condition for somatic cell reprogramming and differentiation of pluripotent stem cells. Reprod Med Biol 2016; 16:58-66. [PMID: 29259452 PMCID: PMC5715877 DOI: 10.1002/rmb2.12011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 09/28/2016] [Indexed: 12/17/2022] Open
Abstract
Aim One of the parameters that greatly affects homeostasis in the body is the pH. Regarding reproductive biology, germ cells, such as oocytes or sperm, are exposed to severe changes in pH, resulting in dramatic changes in their characteristics. To date, the effect of the pH has not been investigated regarding the reprogramming of somatic cells and the maintenance and differentiation of pluripotent stem cells. Methods In order to investigate the effects of the pH on cell culture, the methods to produce induced pluripotent stem cells (iPSCs) and to differentiate embryonic stem cells (ESCs) into mesendoderm and neuroectoderm were performed at each medium pH from 6.6 to 7.8. Using the cells of the Oct4‐GFP (green fluorescent protein) carrying mouse, the effects of pH changes were examined on the timing and colony formation at cell reprogramming and on the cell morphology and direction of the differentiation of the ESCs. Results The colony formation rate and timing of the reprogramming of the somatic cells varied depending on the pH of the culture medium. In addition, mesendodermal differentiation of the mouse ESCs was enhanced at the high pH level of 7.8. Conclusion These results suggest that the pH in the culture medium is one of the key factors in the induction of the reprogramming of somatic cells and in the differentiation of pluripotent stem cells.
Collapse
Affiliation(s)
- Narae Kim
- Laboratory of Reproductive Biology Graduate School of Agriculture Kyoto University Kyoto Japan
| | - Naojiro Minami
- Laboratory of Reproductive Biology Graduate School of Agriculture Kyoto University Kyoto Japan
| | - Masayasu Yamada
- Laboratory of Reproductive Biology Graduate School of Agriculture Kyoto University Kyoto Japan
| | - Hiroshi Imai
- Laboratory of Reproductive Biology Graduate School of Agriculture Kyoto University Kyoto Japan
| |
Collapse
|
16
|
Adler-Wailes DC, Kramer JA, DePamphilis ML. Geminin Is Essential for Pluripotent Cell Viability During Teratoma Formation, but Not for Differentiated Cell Viability During Teratoma Expansion. Stem Cells Dev 2016; 26:285-302. [PMID: 27821018 DOI: 10.1089/scd.2016.0260] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Pluripotent embryonic stem cells (ESCs) are unusual in that geminin has been reported to be essential either to prevent differentiation by maintaining expression of pluripotency genes or to prevent DNA rereplication-dependent apoptosis. To distinguish between these two incompatible hypotheses, immune-compromised mice were inoculated subcutaneously with ESCs harboring conditional Gmnn alleles alone or together with a tamoxifen-dependent Cre recombinase gene. Mice were then injected with tamoxifen at various times during which the ESCs proliferated and differentiated into a teratoma. For comparison, the same ESCs were cultured in vitro in the presence of monohydroxytamoxifen. The results revealed that geminin is a haplosufficient gene that is essential for ESC viability before they differentiate into a teratoma, but once a teratoma is established, the differentiated cells can continue to proliferate in the absence of Gmnn alleles, geminin protein, and pluripotent stem cells. Thus, differentiated cells did not require geminin for efficient proliferation within the context of a solid tissue, although they did when teratoma cells were cultured in vitro. These results provide proof-of-principle that preventing geminin function could prevent malignancy in tumors derived from pluripotent cells by selectively eliminating the progenitor cells with little harm to normal cells.
Collapse
Affiliation(s)
- Diane C Adler-Wailes
- 1 Eunice Kennedy Shriver National Institute of Child Health and Human Development , Bethesda, Maryland
| | - Joshua A Kramer
- 2 Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc. , Bethesda, Maryland
| | - Melvin L DePamphilis
- 1 Eunice Kennedy Shriver National Institute of Child Health and Human Development , Bethesda, Maryland
| |
Collapse
|
17
|
Abstract
The treatment of renal failure has seen little change in the past 70 years. Patients with end-stage renal disease (ESRD) are treated with renal replacement therapy, including dialysis or organ transplantation. The growing imbalance between the availability of donor organs and prevalence of ESRD is pushing an increasing number of patients to undergo dialysis. Although the prospect of new treatment options for patients through regenerative medicine has long been suggested, advances in the generation of human kidney cell types through the directed differentiation of human pluripotent stem cells over the past 2 years have brought this prospect closer to delivery. These advances are the result of careful research into mammalian embryogenesis. By understanding the decision points made within the embryo to pattern the kidney, it is now possible to recreate self-organizing kidney tissues in vitro. In this Review, we describe the key decision points in kidney development and how these decisions have been mimicked experimentally. Recreation of human nephrons from human pluripotent stem cells opens the door to patient-derived disease models and personalized drug and toxicity screening. In the long term, we hope that these efforts will also result in the generation of bioengineered organs for the treatment of kidney disease.
Collapse
|
18
|
Boxman J, Sagy N, Achanta S, Vadigepalli R, Nachman I. Integrated live imaging and molecular profiling of embryoid bodies reveals a synchronized progression of early differentiation. Sci Rep 2016; 6:31623. [PMID: 27530599 PMCID: PMC4987683 DOI: 10.1038/srep31623] [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: 04/25/2016] [Accepted: 07/22/2016] [Indexed: 01/23/2023] Open
Abstract
Embryonic stem cells can spontaneously differentiate into cell types of all germ layers within embryoid bodies (EBs) in a highly variable manner. Whether there exists an intrinsic differentiation program common to all EBs is unknown. Here, we present a novel combination of high-throughput live two-photon imaging and gene expression profiling to study early differentiation dynamics spontaneously occurring within developing EBs. Onset timing of Brachyury-GFP was highly variable across EBs, while the spatial patterns as well as the dynamics of mesendodermal progression following onset were remarkably similar. We therefore defined a 'developmental clock' using the Brachyury-GFP signal onset timing. Mapping snapshot gene expression measurements to this clock revealed their temporal trends, indicating that loss of pluripotency, formation of primitive streak and mesodermal lineage progression are synchronized in EBs. Exogenous activation of Wnt or BMP signaling accelerated the intrinsic clock. CHIR down-regulated Wnt3, allowing insights into dependency mechanisms between canonical Wnt signaling and multiple genes. Our findings reveal a developmental clock characteristic of an early differentiation program common to all EBs, further establishing them as an in vitro developmental model.
Collapse
Affiliation(s)
- Jonathan Boxman
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Israel
| | - Naor Sagy
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Israel
| | - Sirisha Achanta
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rajanikanth Vadigepalli
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Iftach Nachman
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Israel
| |
Collapse
|
19
|
Corritore E, Lee YS, Sokal EM, Lysy PA. β-cell replacement sources for type 1 diabetes: a focus on pancreatic ductal cells. Ther Adv Endocrinol Metab 2016; 7:182-99. [PMID: 27540464 PMCID: PMC4973405 DOI: 10.1177/2042018816652059] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Thorough research on the capacity of human islet transplantation to cure type 1 diabetes led to the achievement of 3- to 5-year-long insulin independence in nearly half of transplanted patients. Yet, translation of this technique to clinical routine is limited by organ shortage and the need for long-term immunosuppression, restricting its use to adults with unstable disease. The production of new bona fide β cells in vitro was thus investigated and finally achieved with human pluripotent stem cells (PSCs). Besides ethical concerns about the use of human embryos, studies are now evaluating the possibility of circumventing the spontaneous tumor formation associated with transplantation of PSCs. These issues fueled the search for cell candidates for β-cell engineering with safe profiles for clinical translation. In vivo studies revealed the regeneration capacity of the exocrine pancreas after injury that depends at least partially on facultative progenitors in the ductal compartment. These stimulated subpopulations of pancreatic ductal cells (PDCs) underwent β-cell transdifferentiation through reactivation of embryonic signaling pathways. In vitro models for expansion and differentiation of purified PDCs toward insulin-producing cells were described using cocktails of growth factors, extracellular-matrix proteins and transcription factor overexpression. In this review, we will describe the latest findings in pancreatic β-cell mass regeneration due to adult ductal progenitor cells. We will further describe recent advances in human PDC transdifferentiation to insulin-producing cells with potential for clinical translational studies.
Collapse
Affiliation(s)
- Elisa Corritore
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium
| | - Yong-Syu Lee
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium
| | - Etienne M. Sokal
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium
| | | |
Collapse
|
20
|
Semrau S, van Oudenaarden A. Studying Lineage Decision-Making In Vitro: Emerging Concepts and Novel Tools. Annu Rev Cell Dev Biol 2015; 31:317-45. [DOI: 10.1146/annurev-cellbio-100814-125300] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | - Alexander van Oudenaarden
- Hubrecht Institute, 3584 CT Utrecht, The Netherlands;
- University Medical Center Utrecht, Cancer Genomics Netherlands, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
21
|
Huang YY, Kaneko KJ, Pan H, DePamphilis ML. Geminin is Essential to Prevent DNA Re-Replication-Dependent Apoptosis in Pluripotent Cells, but not in Differentiated Cells. Stem Cells 2015; 33:3239-53. [PMID: 26140583 DOI: 10.1002/stem.2092] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/11/2015] [Indexed: 01/17/2023]
Abstract
Geminin is a dual-function protein unique to multicellular animals with roles in modulating gene expression and preventing DNA re-replication. Here, we show that geminin is essential at the beginning of mammalian development to prevent DNA re-replication in pluripotent cells, exemplified by embryonic stem cells, as they undergo self-renewal and differentiation. Embryonic stem cells, embryonic fibroblasts, and immortalized fibroblasts were characterized before and after geminin was depleted either by gene ablation or siRNA. Depletion of geminin under conditions that promote either self-renewal or differentiation rapidly induced DNA re-replication, followed by DNA damage, then a DNA damage response, and finally apoptosis. Once differentiation had occurred, geminin was no longer essential for viability, although it continued to contribute to preventing DNA re-replication induced DNA damage. No relationship was detected between expression of geminin and genes associated with either pluripotency or differentiation. Thus, the primary role of geminin at the beginning of mammalian development is to prevent DNA re-replication-dependent apoptosis, a role previously believed essential only in cancer cells. These results suggest that regulation of gene expression by geminin occurs only after pluripotent cells differentiate into cells in which geminin is not essential for viability.
Collapse
Affiliation(s)
- Yi-Yuan Huang
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Kotaro J Kaneko
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Haiyan Pan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Melvin L DePamphilis
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
22
|
Wnt signaling controls the specification of definitive and primitive hematopoiesis from human pluripotent stem cells. Nat Biotechnol 2014; 32:554-61. [PMID: 24837661 PMCID: PMC4152856 DOI: 10.1038/nbt.2915] [Citation(s) in RCA: 331] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 04/23/2014] [Indexed: 12/14/2022]
Abstract
Efforts to derive hematopoietic stem cells (HSCs) from human pluripotent stem cells (hPSCs) are complicated by the fact that embryonic hematopoiesis consists of two programs, primitive and definitive, that differ in developmental potential. As only definitive hematopoiesis generates HSCs, understanding how this program develops is essential for being able to produce this cell population in vitro. Here we show that both hematopoietic programs transition through hemogenic endothelial intermediates and develop from KDR+CD34−CD144− progenitors that are distinguished by CD235a expression. Generation of primitive progenitors (KDR+CD235a+) depends on stage-specific Activin-nodal signaling and inhibition of the Wnt-β-catenin pathway, whereas specification of definitive progenitors (KDR+CD235a−) requires Wnt-β-catenin signaling during this same time frame. Together, these findings establish simple selective differentiation strategies for the generation of primitive or definitive hematopoietic progenitors via Wnt-β-catenin manipulation, and in doing so provide access to enriched populations for future studies on hPSC-derived hematopoietic development.
Collapse
|
23
|
Schiesser JV, Micallef SJ, Hawes S, Elefanty AG, Stanley EG. Derivation of insulin-producing beta-cells from human pluripotent stem cells. Rev Diabet Stud 2014; 11:6-18. [PMID: 25148364 DOI: 10.1900/rds.2014.11.6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Human embryonic stem cells have been advanced as a source of insulin-producing cells that could potentially replace cadaveric-derived islets in the treatment of type 1 diabetes. To this end, protocols have been developed that promote the formation of pancreatic progenitors and endocrine cells from human pluripotent stem cells, encompassing both embryonic stem cells and induced pluripotent stem cells. In this review, we examine these methods and place them in the context of the developmental and embryological studies upon which they are based. In particular, we outline the stepwise differentiation of cells towards definitive endoderm, pancreatic endoderm, endocrine lineages and the emergence of functional beta-cells. In doing so, we identify key factors common to many such protocols and discuss the proposed action of these factors in the context of cellular differentiation and ongoing development. We also compare strategies that entail transplantation of progenitor populations with those that seek to develop fully functional hormone expressing cells in vitro. Overall, our survey of the literature highlights the significant progress already made in the field and identifies remaining deficiencies in developing a pluripotent stem cell based treatment for type 1 diabetes.
Collapse
Affiliation(s)
- Jacqueline V Schiesser
- Monash Immunology and Stem Cell Laboratories (MISCL), Level 3, Building 75, STRIP1, West Ring Road, Monash University, Clayton, Victoria, 3800, Australia
| | - Suzanne J Micallef
- Monash Immunology and Stem Cell Laboratories (MISCL), Level 3, Building 75, STRIP1, West Ring Road, Monash University, Clayton, Victoria, 3800, Australia
| | - Susan Hawes
- Monash Immunology and Stem Cell Laboratories (MISCL), Level 3, Building 75, STRIP1, West Ring Road, Monash University, Clayton, Victoria, 3800, Australia
| | - Andrew G Elefanty
- Monash Immunology and Stem Cell Laboratories (MISCL), Level 3, Building 75, STRIP1, West Ring Road, Monash University, Clayton, Victoria, 3800, Australia
| | - Edouard G Stanley
- Monash Immunology and Stem Cell Laboratories (MISCL), Level 3, Building 75, STRIP1, West Ring Road, Monash University, Clayton, Victoria, 3800, Australia
| |
Collapse
|
24
|
The differentiation of human adipose-derived stem cells towards a urothelium-like phenotype in vitro and the dynamic temporal changes of related cytokines by both paracrine and autocrine signal regulation. PLoS One 2014; 9:e95583. [PMID: 24752317 PMCID: PMC3994076 DOI: 10.1371/journal.pone.0095583] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 03/28/2014] [Indexed: 02/07/2023] Open
Abstract
Purpose To investigate the differentiation ability of human adipose-derived stem cells (ASCs) towards urothelium-like cells in vitro and the dynamic changes of related cytokines and cytokine receptors in the culture medium. Materials and Methods The ASCs were induced using both conditioned media (CM) and the transwell co-culture system with an immortalized urothelium cell line (SV-HUC-1,HUC) for 21 days. Protein and mRNA expression of the mature urothelium specific markers uroplakin-IA (UP-1A) and uroplakin-II (UP-II) were detected by immunofluorescence and quantitative real-time PCR, respectively. Array detection was used to screen 41 cytokines and receptors in the upper medium of urothelium, non-induced ASCs and urothelium-induced ASCs at three time points, early (12 hours), intermediate (7 days) and late (21 days). Results After induction for 7 days, the ASCs grown in both CM and transwell co-culture system expressed uroplakin-IA (13.54±2.00%; 17.28±1.84%) and uroplakin-II (19.49±1.73%; 13.98±1.47%). After induction for 21 days, ASCs grown in co-culture had significantly increased expression of uroplakin-IA (48.03±1.25%; 49.57±2.85%) and uroplakin-II (45.38±2.50%; 46.58±1.95%). In the upper medium of urothelium, 28 cytokines and 8 cytokine receptors had significantly higher expression than the counterpart of non-induced ASCs. After 7 days induction, the expression of 22 cytokines and 8 cytokine receptors was significantly elevated in the upper medium of induced ASCs compared to non-induced ASCs. At the early and intermediate time points, ASCs secreted high levels of relative cytokines and soluble receptors, but their expressions decreased significantly at the late time point. Conclusion The adipose-derived stem cells have the potential to be differentiated into urothelium-like cells in vitro by both CM and transwell co-culture system with mature urothelium. Numerous cytokines and receptors were involved in the differentiation process with dynamic temporal changes by both paracrine and autocrine signal regulation. Further studies should be carried out to determine the detailed mechanism of cytokines and receptors and to enhance the urothelium differentiation efficiency of ASCs.
Collapse
|
25
|
Cimetta E, Vunjak-Novakovic G. Microscale technologies for regulating human stem cell differentiation. Exp Biol Med (Maywood) 2014; 239:1255-63. [PMID: 24737735 DOI: 10.1177/1535370214530369] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
During development and regeneration, tissues emerge from coordinated sequences of stem cell renewal, specialization, and assembly that are orchestrated by cascades of regulatory factors. This complex in vivo milieu, while necessary to fully recapitulate biology and to properly engineer progenitor cells, is difficult to replicate in vitro. We are just starting to fully realize the importance of the entire context of cell microenvironment-the other cells, three-dimensional matrix, molecular and physical signals. Bioengineered environments that combine tissue-specific transport and signaling are critical to study cellular responses at biologically relevant scales and in settings predictive of human condition. We therefore developed microbioreactors that couple the application of fast dynamic changes in environmental signals with versatile, high-throughput operation and imaging capability. Our base device is a microfluidic platform with an array of microwells containing cells or tissue constructs that are exposed to stable concentration gradients. Mathematical modeling of flow and mass transport can predict the shape of these gradients and the kinetic changes in local concentrations. A single platform, the size of a microscope slide, contains up to 120 biological samples. As an example of application, we describe studies of cell fate specification and mesodermal lineage commitment in human embryonic stem cells and induced pluripotent stem cells. The embryoid bodies formed from these cells were subjected to single and multiple concentration gradients of Wnt3a, Activin A, bone morphogenic protein 4 (BMP4), and their inhibitors, and the gene expression profiles were correlated to the concentration gradients of morphogens to identify the exact conditions for mesodermal differentiation.
Collapse
Affiliation(s)
- Elisa Cimetta
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | | |
Collapse
|
26
|
Hughes JN, Wong CKE, Lau KX, Rathjen PD, Rathjen J. Regulation of pluripotent cell differentiation by a small molecule, staurosporine. Differentiation 2014; 87:101-10. [PMID: 24582574 DOI: 10.1016/j.diff.2014.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 12/16/2013] [Accepted: 01/07/2014] [Indexed: 12/25/2022]
Abstract
Research in the embryo and in culture has resulted in a sophisticated understanding of many regulators of pluripotent cell differentiation. As a consequence, protocols for the differentiation of pluripotent cells generally rely on a combination of exogenous growth factors and endogenous signalling. Little consideration has been given to manipulating other pathways to achieve pluripotent cell differentiation. The integrity of cell:cell contacts has been shown to influence lineage choice during pluripotent cell differentiation, with disruption of cell:cell contacts promoting mesendoderm formation and maintenance of cell:cell contacts resulting in the preferential formation of neurectoderm. Staurosporine is a broad spectrum inhibitor of serine/threonine kinases which has several effects on cell function, including interruption of cell:cell contacts, decreasing focal contact size, inducing epithelial to mesenchyme transition (EMT) and promoting cell differentiation. The possibility that staurosporine could influence lineage choice from pluripotent cells in culture was investigated. The addition of staurosporine to differentiating mouse EPL resulted in preferential formation of mesendoderm and mesoderm populations, and inhibited the formation of neurectoderm. Addition of staurosporine to human ES cells similarly induced primitive streak marker gene expression. These data demonstrate the ability of staurosporine to influence lineage choice during pluripotent cell differentiation and to mimic the effect of disrupting cell:cell contacts. Staurosporine induced mesendoderm in the absence of known inducers of formation, such as serum and BMP4. Staurosporine induced the expression of mesendoderm markers, including markers that were not induced by BMP4, suggesting it acted as a broad spectrum inducer of molecular gastrulation. This approach has identified a small molecule regulator of lineage choice with potential applications in the commercial development of ES cell derivatives, specifically as a method for forming mesendoderm progenitors or as a culture adjunct to prevent the formation of ectoderm progenitors during pluripotent cell differentiation.
Collapse
Affiliation(s)
- James Nicholas Hughes
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Chong Kum Edwin Wong
- Department of Zoology, University of Melbourne, Parkville, Victoria 3010 Australia; Australian Stem Cell Centre, Monash University, Clayton, 3800 Victoria, Australia
| | - Kevin Xiuwen Lau
- Department of Zoology, University of Melbourne, Parkville, Victoria 3010 Australia
| | - Peter David Rathjen
- Department of Zoology, University of Melbourne, Parkville, Victoria 3010 Australia; The Menzies Research Institute Tasmania, University of Tasmania, 17 Liverpool Street, Hobart, Tasmania 7000, Australia.
| | - Joy Rathjen
- Department of Zoology, University of Melbourne, Parkville, Victoria 3010 Australia; The Menzies Research Institute Tasmania, University of Tasmania, 17 Liverpool Street, Hobart, Tasmania 7000, Australia.
| |
Collapse
|
27
|
Schiesser JV, Wells JM. Generation of β cells from human pluripotent stem cells: are we there yet? Ann N Y Acad Sci 2014; 1311:124-37. [PMID: 24611778 DOI: 10.1111/nyas.12369] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In 1998, the landmark paper describing the isolation and culture of human embryonic stem cells (ESCs) was published. Since that time, the main goal of many diabetes researchers has been to derive β cells from ESCs as a renewable cell-based therapy for the treatment of patients with diabetes. In working toward this goal, numerous protocols that attempt to recapitulate normal pancreatic development have been published that result in the formation of pancreatic cell types from human pluripotent cells. This review examines stem cell differentiation methods and places them within the context of pancreatic development. We additionally compare strategies that are currently being used to generate pancreatic cell types and contrast them with approaches that have been used to generate functional cell types in different lineages. In doing this, we aim to identify how new approaches might be used to improve yield and functionality of in vitro-derived pancreatic β cells as an eventual cell-based therapy for type 1 diabetes.
Collapse
Affiliation(s)
- Jacqueline V Schiesser
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | |
Collapse
|
28
|
Kinney MA, Hookway TA, Wang Y, McDevitt TC. Engineering three-dimensional stem cell morphogenesis for the development of tissue models and scalable regenerative therapeutics. Ann Biomed Eng 2014; 42:352-67. [PMID: 24297495 PMCID: PMC3939035 DOI: 10.1007/s10439-013-0953-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/21/2013] [Indexed: 12/11/2022]
Abstract
The physiochemical stem cell microenvironment regulates the delicate balance between self-renewal and differentiation. The three-dimensional assembly of stem cells facilitates cellular interactions that promote morphogenesis, analogous to the multicellular, heterotypic tissue organization that accompanies embryogenesis. Therefore, expansion and differentiation of stem cells as multicellular aggregates provides a controlled platform for studying the biological and engineering principles underlying spatiotemporal morphogenesis and tissue patterning. Moreover, three-dimensional stem cell cultures are amenable to translational screening applications and therapies, which underscores the broad utility of scalable suspension cultures across laboratory and clinical scales. In this review, we discuss stem cell morphogenesis in the context of fundamental biophysical principles, including the three-dimensional modulation of adhesions, mechanics, and molecular transport and highlight the opportunities to employ stem cell spheroids for tissue modeling, bioprocessing, and regenerative therapies.
Collapse
Affiliation(s)
- Melissa A. Kinney
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology/Emory University, Atlanta, GA, USA
| | - Tracy A. Hookway
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology/Emory University, Atlanta, GA, USA
| | - Yun Wang
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology/Emory University, Atlanta, GA, USA
| | - Todd C. McDevitt
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology/Emory University, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
29
|
Yap C, Goh HN, Familari M, Rathjen PD, Rathjen J. The formation of proximal and distal definitive endoderm populations in culture requires p38 MAPK activity. J Cell Sci 2014; 127:2204-16. [PMID: 24481813 DOI: 10.1242/jcs.134502] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Endoderm formation in the mammal is a complex process with two lineages forming during the first weeks of development, the primitive (or extraembryonic) endoderm, which is specified in the blastocyst, and the definitive endoderm that forms later, at gastrulation, as one of the germ layers of the embryo proper. Fate mapping evidence suggests that the definitive endoderm arises as two waves, which potentially reflect two distinct cell populations. Early primitive ectoderm-like (EPL) cell differentiation has been used successfully to identify and characterise mechanisms regulating molecular gastrulation and lineage choice during differentiation. The roles of the p38 MAPK family in the formation of definitive endoderm were investigated using EPL cells and chemical inhibitors of p38 MAPK activity. These approaches define a role for p38 MAPK activity in the formation of the primitive streak and a second role in the formation of the definitive endoderm. Characterisation of the definitive endoderm populations formed from EPL cells demonstrates the formation of two distinct populations, defined by gene expression and ontogeny, that were analogous to the proximal and distal definitive endoderm populations of the embryo. Formation of the proximal definitive endoderm was found to require p38 MAPK activity and is correlated with molecular gastrulation, defined by the expression of brachyury (T). Distal definitive endoderm formation also requires p38 MAPK activity but can form when T expression is inhibited. Understanding lineage complexity will be a prerequisite for the generation of endoderm derivatives for commercial and clinical use.
Collapse
Affiliation(s)
- Charlotte Yap
- Department of Zoology, University of Melbourne, Victoria, 3010, Australia
| | - Hwee Ngee Goh
- Department of Zoology, University of Melbourne, Victoria, 3010, Australia
| | - Mary Familari
- Department of Zoology, University of Melbourne, Victoria, 3010, Australia
| | - Peter David Rathjen
- Department of Zoology, University of Melbourne, Victoria, 3010, Australia The Menzies Research Institute Tasmania, University of Tasmania, Tasmania, 7000, Australia
| | - Joy Rathjen
- Department of Zoology, University of Melbourne, Victoria, 3010, Australia The Menzies Research Institute Tasmania, University of Tasmania, Tasmania, 7000, Australia
| |
Collapse
|
30
|
Sangel P, Oka M, Yoneda Y. The role of Importin-βs in the maintenance and lineage commitment of mouse embryonic stem cells. FEBS Open Bio 2014; 4:112-20. [PMID: 24490135 PMCID: PMC3907685 DOI: 10.1016/j.fob.2014.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 11/29/2013] [Accepted: 01/02/2014] [Indexed: 01/08/2023] Open
Abstract
Members of the Importin-β family recognize nuclear localization signals (NLS) and nuclear export signals (NES). These proteins play important roles in various nucleocytoplasmic transport processes in cells. Here, we examined the expression patterns of 21 identified Importin-β genes in mouse embryonic stem cells (mESCs), mouse embryonic fibroblast (MEF) and mESCs differentiated into neural ectoderm (NE) or mesoendoderm (ME). We observed striking differences in the Importin-β mRNA expression levels within these cell types. We also found that knockdown of selected Importin-β genes led to suppression of Nanog, and altered the balance of Oct4/Sox2 expression ratio, which is important for NE/ME lineage choice. Furthermore, we demonstrated that knockdown of XPO4, RanBP17, RanBP16, or IPO7 differentially affected the lineage selection of differentiating mESCs. More specifically, knockdown of XPO4 selectively stimulated the mESC differentiation towards definitive endoderm, while concomitantly inhibiting NE differentiation. RanBP17 knockdown also promoted endodermal differentiation with no effect on NE differentiation. RanBP16 knockdown caused differentiation into ME, while IPO7 knockdown inhibited NE differentiation, without obvious effects on the other lineages. Collectively, our results suggest that Importin-βs play important roles in cell fate determination processes of mESCs, such as in the maintenance of pluripotency or selection of lineage during differentiation. Importin-β expression patterns are distinct in mESCs, MEFs, NE and ME cells. Importin-β may modulate differentiation and lineage selection in mESCs. Suppression of either XPO4 or RanBP17 induces endodermal differentiation in mESCs. RanBP16 suppression induces a ME differentiation in mESCs. XPO4 and IPO7 are essential for mESC differentiation into NE cells.
Collapse
Affiliation(s)
- Percival Sangel
- Biomolecular Dynamics Laboratory, Department of Frontier Bioscience, Graduate School of Frontier Bioscience, Osaka University, 1-3 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Masahiro Oka
- Biomolecular Dynamics Laboratory, Department of Frontier Bioscience, Graduate School of Frontier Bioscience, Osaka University, 1-3 Yamada-oka, Suita, Osaka 565-0871, Japan ; Department of Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan ; JST, CREST, Osaka University, 1-3 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Yoshihiro Yoneda
- Biomolecular Dynamics Laboratory, Department of Frontier Bioscience, Graduate School of Frontier Bioscience, Osaka University, 1-3 Yamada-oka, Suita, Osaka 565-0871, Japan ; Department of Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan ; JST, CREST, Osaka University, 1-3 Yamada-oka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
31
|
Lam AQ, Freedman BS, Morizane R, Lerou PH, Valerius MT, Bonventre JV. Rapid and efficient differentiation of human pluripotent stem cells into intermediate mesoderm that forms tubules expressing kidney proximal tubular markers. J Am Soc Nephrol 2013; 25:1211-25. [PMID: 24357672 DOI: 10.1681/asn.2013080831] [Citation(s) in RCA: 209] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) can generate a diversity of cell types, but few methods have been developed to derive cells of the kidney lineage. Here, we report a highly efficient system for differentiating human embryonic stem cells and induced pluripotent stem cells (referred to collectively as hPSCs) into cells expressing markers of the intermediate mesoderm (IM) that subsequently form tubule-like structures. Treatment of hPSCs with the glycogen synthase kinase-3β inhibitor CHIR99021 induced BRACHYURY(+)MIXL1(+) mesendoderm differentiation with nearly 100% efficiency. In the absence of additional exogenous factors, CHIR99021-induced mesendodermal cells preferentially differentiated into cells expressing markers of lateral plate mesoderm with minimal IM differentiation. However, the sequential treatment of hPSCs with CHIR99021 followed by fibroblast growth factor-2 and retinoic acid generated PAX2(+)LHX1(+) cells with 70%-80% efficiency after 3 days of differentiation. Upon growth factor withdrawal, these PAX2(+)LHX1(+) cells gave rise to apically ciliated tubular structures that coexpressed the proximal tubule markers Lotus tetragonolobus lectin, N-cadherin, and kidney-specific protein and partially integrated into embryonic kidney explant cultures. With the addition of FGF9 and activin, PAX2(+)LHX1(+) cells specifically differentiated into cells expressing SIX2, SALL1, and WT1, markers of cap mesenchyme nephron progenitor cells. Our findings demonstrate the effective role of fibroblast growth factor signaling in inducing IM differentiation in hPSCs and establish the most rapid and efficient system whereby hPSCs can be differentiated into cells with features characteristic of kidney lineage cells.
Collapse
Affiliation(s)
- Albert Q Lam
- Renal Division, Department of Medicine, and Harvard Stem Cell Institute, Cambridge, Massachusetts; and
| | - Benjamin S Freedman
- Renal Division, Department of Medicine, and Harvard Stem Cell Institute, Cambridge, Massachusetts; and
| | - Ryuji Morizane
- Renal Division, Department of Medicine, and Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Paul H Lerou
- Harvard Stem Cell Institute, Cambridge, Massachusetts; and Department of Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - M Todd Valerius
- Renal Division, Department of Medicine, and Harvard Stem Cell Institute, Cambridge, Massachusetts; and
| | - Joseph V Bonventre
- Renal Division, Department of Medicine, and Harvard Stem Cell Institute, Cambridge, Massachusetts; and
| |
Collapse
|
32
|
Abstract
Autism and autism spectrum disorders (ASDs) are complex neurodevelopmental disorders. ASDs are clinically defined by deficits in communication, social skills, and repetitive and/or restrictive interests and behaviours. With the prevalence rates for ASDs rapidly increasing, the need for effective therapies for autism is a priority for biomedical research. Currently available medications do not target the core symptoms, can have markedly adverse side-effects, and are mainly palliative for negative behaviours. The development of molecular and regenerative interventions is progressing rapidly, and medicine holds great expectations for stem cell therapies. Cells could be designed to target the observed molecular mechanisms of ASDs, that is, abnormal neurotransmitter regulation, activated microglia, mitochondrial dysfunction, blood-brain barrier disruptions, and chronic intestinal inflammation. Presently, the paracrine, secretome, and immunomodulatory effects of stem cells would appear to be the likely mechanisms of application for ASD therapeutics. This review will focus on the potential use of the various types of stem cells: embryonic, induced pluripotential, fetal, and adult stem cells as targets for ASD therapeutics.
Collapse
|
33
|
Perspectives on the use of stem cells for autism treatment. Stem Cells Int 2013; 2013:262438. [PMID: 24222772 PMCID: PMC3810518 DOI: 10.1155/2013/262438] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 08/22/2013] [Accepted: 09/06/2013] [Indexed: 12/13/2022] Open
Abstract
Autism and autism spectrum disorders (ASDs) are complex neurodevelopmental disorders. ASDs are clinically defined by deficits in communication, social skills, and repetitive and/or restrictive interests and behaviours. With the prevalence rates for ASDs rapidly increasing, the need for effective therapies for autism is a priority for biomedical research. Currently available medications do not target the core symptoms, can have markedly adverse side-effects, and are mainly palliative for negative behaviours. The development of molecular and regenerative interventions is progressing rapidly, and medicine holds great expectations for stem cell therapies. Cells could be designed to target the observed molecular mechanisms of ASDs, that is, abnormal neurotransmitter regulation, activated microglia, mitochondrial dysfunction, blood-brain barrier disruptions, and chronic intestinal inflammation. Presently, the paracrine, secretome, and immunomodulatory effects of stem cells would appear to be the likely mechanisms of application for ASD therapeutics. This review will focus on the potential use of the various types of stem cells: embryonic, induced pluripotential, fetal, and adult stem cells as targets for ASD therapeutics.
Collapse
|
34
|
WNT3A promotes hematopoietic or mesenchymal differentiation from hESCs depending on the time of exposure. Stem Cell Reports 2013; 1:53-65. [PMID: 24052942 PMCID: PMC3757745 DOI: 10.1016/j.stemcr.2013.04.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 04/21/2013] [Accepted: 04/22/2013] [Indexed: 11/22/2022] Open
Abstract
We investigated the role of canonical WNT signaling in mesoderm and hematopoietic development from human embryonic stem cells (hESCs) using a recombinant human protein-based differentiation medium (APEL). In contrast to prior studies using less defined culture conditions, we found that WNT3A alone was a poor inducer of mesoderm. However, WNT3A synergized with BMP4 to accelerate mesoderm formation, increase embryoid body size, and increase the number of hematopoietic blast colonies. Interestingly, inclusion of WNT3A or a GSK3 inhibitor in methylcellulose colony-forming assays at 4 days of differentiation abrogated blast colony formation but supported the generation of mesospheres that expressed genes associated with mesenchymal lineages. Mesospheres differentiated into cells with characteristics of bone, fat, and smooth muscle. These studies identify distinct effects for WNT3A, supporting the formation of hematopoietic or mesenchymal lineages from human embryonic stem cells, depending upon differentiation stage at the time of exposure.
Collapse
|
35
|
Faunes F, Hayward P, Descalzo SM, Chatterjee SS, Balayo T, Trott J, Christoforou A, Ferrer-Vaquer A, Hadjantonakis AK, Dasgupta R, Arias AM. A membrane-associated β-catenin/Oct4 complex correlates with ground-state pluripotency in mouse embryonic stem cells. Development 2013; 140:1171-83. [PMID: 23444350 PMCID: PMC3585656 DOI: 10.1242/dev.085654] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The maintenance of pluripotency in mouse embryonic stem cells (mESCs) relies on the activity of a transcriptional network that is fuelled by the activity of three transcription factors (Nanog, Oct4 and Sox2) and balanced by the repressive activity of Tcf3. Extracellular signals modulate the activity of the network and regulate the differentiation capacity of the cells. Wnt/β-catenin signaling has emerged as a significant potentiator of pluripotency: increases in the levels of β-catenin regulate the activity of Oct4 and Nanog, and enhance pluripotency. A recent report shows that β-catenin achieves some of these effects by modulating the activity of Tcf3, and that this effect does not require its transcriptional activation domain. Here, we show that during self-renewal there is negligible transcriptional activity of β-catenin and that this is due to its tight association with membranes, where we find it in a complex with Oct4 and E-cadherin. Differentiation triggers a burst of Wnt/β-catenin transcriptional activity that coincides with the disassembly of the complex. Our results establish that β-catenin, but not its transcriptional activity, is central to pluripotency acting through a β-catenin/Oct4 complex.
Collapse
Affiliation(s)
- Fernando Faunes
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Cimetta E, Sirabella D, Yeager K, Davidson K, Simon J, Moon RT, Vunjak-Novakovic G. Microfluidic bioreactor for dynamic regulation of early mesodermal commitment in human pluripotent stem cells. LAB ON A CHIP 2013; 13:355-64. [PMID: 23232509 PMCID: PMC3535552 DOI: 10.1039/c2lc40836h] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
During development and regeneration, tissues emerge from coordinated sequences of stem cell renewal, specialization and assembly that are orchestrated by cascades of regulatory signals. The complex and dynamic in vivo milieu cannot be replicated using standard in vitro techniques. Microscale technologies now offer potential for conducting highly controllable and sophisticated experiments at biologically relevant scales, with real-time insights into cellular responses. We developed a microbioreactor providing time sequences of space-resolved gradients of multiple molecular factors in three-dimensional (3D) cell culture settings, along with a versatile, high-throughput operation and imaging compatibility. A single microbioreactor yields up to 120 data points, corresponding to 15 replicates of a gradient with 8 concentration levels. Embryoid bodies (EBs) obtained from human embryonic and induced pluripotent stem cells (hESC, hiPSC) were exposed to concentration gradients of Wnt3a, Activin A, BMP4 and their inhibitors, to get new insights into the early-stage fate specification and mesodermal lineage commitment. We were able to evaluate the initiation of mesodermal induction by measuring and correlating the gene expression profiles to the concentration gradients of mesoderm-inducing morphogens. We propose that the microbioreactor systems combining spatial and temporal gradients of molecular and physical factors to hESC and hiPSC cultures can form a basis for predictable in vitro models of development and disease.
Collapse
Affiliation(s)
- Elisa Cimetta
- Columbia University, Department of Biomedical Engineering, Vanderbilt Clinic, New York, NY 10032, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Titmarsh DM, Hudson JE, Hidalgo A, Elefanty AG, Stanley EG, Wolvetang EJ, Cooper-White JJ. Microbioreactor arrays for full factorial screening of exogenous and paracrine factors in human embryonic stem cell differentiation. PLoS One 2012; 7:e52405. [PMID: 23300662 PMCID: PMC3530582 DOI: 10.1371/journal.pone.0052405] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Accepted: 11/14/2012] [Indexed: 02/06/2023] Open
Abstract
Timed exposure of pluripotent stem cell cultures to exogenous molecules is widely used to drive differentiation towards desired cell lineages. However, screening differentiation conditions in conventional static cultures can become impractical in large parameter spaces, and is intrinsically limited by poor spatiotemporal control of the microenvironment that also makes it impossible to determine whether exogenous factors act directly or through paracrine-dependent mechanisms. We detail here the development of a continuous flow microbioreactor array platform that combines full-factorial multiplexing of input factors with progressive accumulation of paracrine factors through serially-connected culture chambers, and further, the use of this system to explore the combinatorial parameter space of both exogenous and paracrine factors involved in human embryonic stem cell (hESC) differentiation to a MIXL1-GFP(+) primitive streak-like population. We show that well known inducers of primitive streak (BMP, Activin and Wnt signals) do not simply act directly on hESC to induce MIXL1 expression, but that this requires accumulation of surplus, endogenous factors; and, that conditioned medium or FGF-2 supplementation is able to offset this. Our approach further reveals the presence of a paracrine, negative feedback loop to the MIXL1-GFP(+) population, which can be overcome with GSK-3β inhibitors (BIO or CHIR99021), implicating secreted Wnt inhibitory signals such as DKKs and sFRPs as candidate effectors. Importantly, modulating paracrine effects identified in microbioreactor arrays by supplementing FGF-2 and CHIR in conventional static culture vessels resulted in improved differentiation outcomes. We therefore demonstrate that this microbioreactor array platform uniquely enables the identification and decoding of complex soluble factor signalling hierarchies, and that this not only challenges prevailing strategies for extrinsic control of hESC differentiation, but also is translatable to conventional culture systems.
Collapse
Affiliation(s)
- Drew M. Titmarsh
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St. Lucia, Queensland, Australia
| | - James E. Hudson
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St. Lucia, Queensland, Australia
| | - Alejandro Hidalgo
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St. Lucia, Queensland, Australia
| | - Andrew G. Elefanty
- Murdoch Childrens Research Institute, The Royal Children’s Hospital, Flemington Road, Parkville, Victoria, Australia
- Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, Victoria, Australia
| | - Edouard G. Stanley
- Murdoch Childrens Research Institute, The Royal Children’s Hospital, Flemington Road, Parkville, Victoria, Australia
- Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, Victoria, Australia
| | - Ernst J. Wolvetang
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St. Lucia, Queensland, Australia
| | - Justin J. Cooper-White
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St. Lucia, Queensland, Australia
- School of Chemical Engineering, The University of Queensland, St. Lucia, Queensland, Australia
- * E-mail:
| |
Collapse
|
38
|
Teo AKK, Ali Y, Wong KY, Chipperfield H, Sadasivam A, Poobalan Y, Tan EK, Wang ST, Abraham S, Tsuneyoshi N, Stanton LW, Dunn NR. Activin and BMP4 synergistically promote formation of definitive endoderm in human embryonic stem cells. Stem Cells 2012; 30:631-42. [PMID: 22893457 DOI: 10.1002/stem.1022] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human embryonic stem cells (hESCs) herald tremendous promise for the production of clinically useful cell types for the treatment of injury and disease. Numerous reports demonstrate their differentiation into definitive endoderm (DE) cells, the germ layer from which pancreatic β cells and hepatocytes arise, solely from exposure to a high dose of recombinant Activin/Nodal. We show that combining a second related ligand, BMP4, in combination with Activin A yields 15%-20% more DE as compared with Activin A alone. The addition of recombinant BMP4 accelerates the downregulation of pluripotency genes, particularly SOX2, and results in upregulation of endogenous BMP2 and BMP4, which in turn leads to elevated levels of phospho-SMAD1/5/8. Combined Activin A and BMP4 treatment also leads to an increase in the expression of DE genes CXCR4, SOX17, and FOXA2 when compared with Activin A addition alone. Comparative microarray studies between DE cells harvested on day 3 of differentiation further reveal a novel set of genes upregulated in response to initial BMP4 exposure. Several of these, including APLNR, LRIG3, MCC, LEPREL1, ROR2, and LZTS1, are expressed in the mouse primitive streak, the site of DE formation. Thus, this synergism between Activin A and BMP4 during the in vitro differentiation of hESC into DE suggests a complex interplay between BMP and Activin/Nodal signaling during the in vivo allocation and expansion of the endoderm lineage.
Collapse
Affiliation(s)
- Adrian K K Teo
- Institute of Medical Biology, Agency for Science, Technology and Research, #06-06 Immunos, Singapore 138648
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
T lymphocyte potential marks the emergence of definitive hematopoietic progenitors in human pluripotent stem cell differentiation cultures. Cell Rep 2012; 2:1722-35. [PMID: 23219550 DOI: 10.1016/j.celrep.2012.11.003] [Citation(s) in RCA: 318] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 11/02/2012] [Accepted: 11/07/2012] [Indexed: 12/13/2022] Open
Abstract
The efficient generation of hematopoietic stem cells from human pluripotent stem cells is dependent on the appropriate specification of the definitive hematopoietic program during differentiation. In this study, we used T lymphocyte potential to track the onset of definitive hematopoiesis from human embryonic and induced pluripotent stem cells differentiated with specific morphogens in serum- and stromal-free cultures. We show that this program develops from a progenitor population with characteristics of hemogenic endothelium, including the expression of CD34, VE-cadherin, GATA2, LMO2, and RUNX1. Along with T cells, these progenitors display the capacity to generate myeloid and erythroid cells. Manipulation of Activin/Nodal signaling during early stages of differentiation revealed that development of the definitive hematopoietic progenitor population is not dependent on this pathway, distinguishing it from primitive hematopoiesis. Collectively, these findings demonstrate that it is possible to generate T lymphoid progenitors from pluripotent stem cells and that this lineage develops from a population whose emergence marks the onset of human definitive hematopoiesis.
Collapse
|
40
|
Minami I, Yamada K, Otsuji TG, Yamamoto T, Shen Y, Otsuka S, Kadota S, Morone N, Barve M, Asai Y, Tenkova-Heuser T, Heuser JE, Uesugi M, Aiba K, Nakatsuji N. A small molecule that promotes cardiac differentiation of human pluripotent stem cells under defined, cytokine- and xeno-free conditions. Cell Rep 2012; 2:1448-60. [PMID: 23103164 DOI: 10.1016/j.celrep.2012.09.015] [Citation(s) in RCA: 185] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 05/18/2012] [Accepted: 09/12/2012] [Indexed: 12/15/2022] Open
Abstract
Human pluripotent stem cells (hPSCs), including embryonic stem cells and induced pluripotent stem cells, are potentially useful in regenerative therapies for heart disease. For medical applications, clinical-grade cardiac cells must be produced from hPSCs in a defined, cost-effective manner. Cell-based screening led to the discovery of KY02111, a small molecule that promotes differentiation of hPSCs to cardiomyocytes. Although the direct target of KY02111 remains unknown, results of the present study suggest that KY02111 promotes differentiation by inhibiting WNT signaling in hPSCs but in a manner that is distinct from that of previously studied WNT inhibitors. Combined use of KY02111 and WNT signaling modulators produced robust cardiac differentiation of hPSCs in a xeno-free, defined medium, devoid of serum and any kind of recombinant cytokines and hormones, such as BMP4, Activin A, or insulin. The methodology has potential as a means for the practical production of human cardiomyocytes for regeneration therapies.
Collapse
Affiliation(s)
- Itsunari Minami
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Pereira LA, Wong MS, Mei Lim S, Stanley EG, Elefanty AG. The Mix family of homeobox genes—Key regulators of mesendoderm formation during vertebrate development. Dev Biol 2012; 367:163-77. [DOI: 10.1016/j.ydbio.2012.04.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 04/24/2012] [Accepted: 04/30/2012] [Indexed: 10/28/2022]
|
42
|
Kim PTW, Ong CJ. Differentiation of definitive endoderm from mouse embryonic stem cells. Results Probl Cell Differ 2012; 55:303-19. [PMID: 22918814 DOI: 10.1007/978-3-642-30406-4_17] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Efficient production of definitive endoderm from embryonic stem (ES) cells opens doors to the possibilities of differentiation of endoderm-derived tissues such as the intestines, pancreas, and liver that could address the needs of people with chronic diseases involving these organs. The lessons learned from developmental biology have contributed significantly to in vitro differentiation of definitive endoderm. Gastrulation, a process that results in the production of all three embryonic germ cell layers, definitive endoderm, mesoderm, and ectoderm, is an important step in embryonic development. Gastrulation occurs as a result of the events that are orchestrated by the signaling pathways involving Nodal, FGF, Wnt, and BMP. Understanding these signaling pathways has led to the introduction of key ingredients such as Activin A, FGF, Wnt, and BMP to the differentiation protocols that have been able to produce definitive endoderm from ES cells. Efficient production of definitive endoderm needs to meet the specific criteria that include (a) increase in the production of markers of definitive endoderm such as Sox 17, FOXA2, GSC, and Mixl1; (b) decrease in the production of markers of primitive/visceral/parietal endoderm, Sox 7 and OCT4; and (c) decrease in the mesoderm markers (Brachyury, MEOX) and ectoderm markers (Sox1 and ZIC1).
Collapse
Affiliation(s)
- Peter T W Kim
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
43
|
Przybyla L, Voldman J. Probing embryonic stem cell autocrine and paracrine signaling using microfluidics. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2012; 5:293-315. [PMID: 22524217 PMCID: PMC4030416 DOI: 10.1146/annurev-anchem-062011-143122] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Although stem cell fate is traditionally manipulated by exogenously altering the cells' extracellular signaling environment, the endogenous autocrine and paracrine signals produced by the cells also contribute to their two essential processes: self-renewal and differentiation. Autocrine and/or paracrine signals are fundamental to both embryonic stem cell self-renewal and early embryonic development, but the nature and contributions of these signals are often difficult to fully define using conventional methods. Microfluidic techniques have been used to explore the effects of cell-secreted signals by controlling cell organization or by providing precise control over the spatial and temporal cellular microenvironment. Here we review how such techniques have begun to be adapted for use with embryonic stem cells, and we illustrate how many remaining questions in embryonic stem cell biology could be addressed using microfluidic technologies.
Collapse
Affiliation(s)
- Laralynne Przybyla
- Dept. of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, USA, 02139
| | - Joel Voldman
- Dept. Of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, USA, 02139
| |
Collapse
|
44
|
Hudson J, Titmarsh D, Hidalgo A, Wolvetang E, Cooper-White J. Primitive cardiac cells from human embryonic stem cells. Stem Cells Dev 2011; 21:1513-23. [PMID: 21933026 DOI: 10.1089/scd.2011.0254] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pluripotent stem cell-derived cardiomyocytes are currently being investigated for in vitro human heart models and as potential therapeutics for heart failure. In this study, we have developed a differentiation protocol that minimizes the need for specific human embryonic stem cell (hESC) line optimization. We first reduced the heterogeneity that exists within the starting population of bulk cultured hESCs by using cells adapted to single-cell passaging in a 2-dimensional (2D) culture format. Compared with bulk cultures, single-cell cultures comprised larger fractions of TG30(hi)/OCT4(hi) cells, corresponding to an increased expression of pluripotency markers OCT4 and NANOG, and reduced expression of early lineage-specific markers. A 2D temporal differentiation protocol was then developed, aimed at reducing the inherent heterogeneity and variability of embryoid body-based protocols, with induction of primitive streak cells using bone morphogenetic protein 4 and activin A, followed by cardiogenesis via inhibition of Wnt signaling using the small molecules IWP-4 or IWR-1. IWP-4 treatment resulted in a large percentage of cells expressing low amounts of cardiac myosin heavy chain and expression of early cardiac progenitor markers ISL1 and NKX2-5, thus indicating the production of large numbers of immature cardiomyocytes (~65,000/cm(2) or ~1.5 per input hESC). This protocol was shown to be effective in HES3, H9, and, to a lesser, extent, MEL1 hESC lines. In addition, we observed that IWR-1 induced predominantly atrial myosin light chain (MLC2a) expression, whereas IWP-4 induced expression of both atrial (MLC2a) and ventricular (MLC2v) forms. The intrinsic flexibility and scalability of this 2D protocol mean that the output population of primitive cardiomyocytes will be particularly accessible and useful for the investigation of molecular mechanisms driving terminal cardiomyocyte differentiation, and potentially for the future treatment of heart failure.
Collapse
Affiliation(s)
- James Hudson
- Tissue Engineering and Microfluidics Group, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Australia
| | | | | | | | | |
Collapse
|
45
|
Pluripotency factors in embryonic stem cells regulate differentiation into germ layers. Cell 2011; 145:875-89. [PMID: 21663792 DOI: 10.1016/j.cell.2011.05.017] [Citation(s) in RCA: 419] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 03/03/2011] [Accepted: 05/16/2011] [Indexed: 02/06/2023]
Abstract
Cell fate decisions are fundamental for development, but we do not know how transcriptional networks reorganize during the transition from a pluripotent to a differentiated cell state. Here, we asked how mouse embryonic stem cells (ESCs) leave the pluripotent state and choose between germ layer fates. By analyzing the dynamics of the transcriptional circuit that maintains pluripotency, we found that Oct4 and Sox2, proteins that maintain ESC identity, also orchestrate germ layer fate selection. Oct4 suppresses neural ectodermal differentiation and promotes mesendodermal differentiation; Sox2 inhibits mesendodermal differentiation and promotes neural ectodermal differentiation. Differentiation signals continuously and asymmetrically modulate Oct4 and Sox2 protein levels, altering their binding pattern in the genome, and leading to cell fate choice. The same factors that maintain pluripotency thus also integrate external signals and control lineage selection. Our study provides a framework for understanding how complex transcription factor networks control cell fate decisions in progenitor cells.
Collapse
|
46
|
Abstract
Embryonic stem cell (ESC) pluripotency is maintained by core transcriptional circuits whereby critical factors sustain their own expression while preventing the expression of genes required for differentiation. Thomson et al. (2011) now show that two core components of the pluripotency circuit, Oct4 and Sox2, are also critical for germ layer fate choice.
Collapse
Affiliation(s)
- Nicola Iovino
- Institut de Génétique Humaine, CNRS, 141 rue de la Cardonille, 34396 Montpellier, France
| | | |
Collapse
|
47
|
Xu X, Browning VL, Odorico JS. Activin, BMP and FGF pathways cooperate to promote endoderm and pancreatic lineage cell differentiation from human embryonic stem cells. Mech Dev 2011; 128:412-27. [PMID: 21855631 DOI: 10.1016/j.mod.2011.08.001] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 08/01/2011] [Accepted: 08/04/2011] [Indexed: 10/17/2022]
Abstract
The study of how human embryonic stem cells (hESCs) differentiate into insulin-producing beta cells has twofold significance: first, it provides an in vitro model system for the study of human pancreatic development, and second, it serves as a platform for the ultimate production of beta cells for transplantation into patients with diabetes. The delineation of growth factor interactions regulating pancreas specification from hESCs in vitro is critical to achieving these goals. In this study, we describe the roles of growth factors bFGF, BMP4 and Activin A in early hESC fate determination. The entire differentiation process is carried out in serum-free chemically-defined media (CDM) and results in reliable and robust induction of pancreatic endoderm cells, marked by PDX1, and cell clusters co-expressing markers characteristic of beta cells, including PDX1 and insulin/C-peptide. Varying the combinations of growth factors, we found that treatment of hESCs with bFGF, Activin A and BMP4 (FAB) together for 3-4days resulted in strong induction of primitive-streak and definitive endoderm-associated genes, including MIXL1, GSC, SOX17 and FOXA2. Early proliferative foregut endoderm and pancreatic lineage cells marked by PDX1, FOXA2 and SOX9 expression are specified in EBs made from FAB-treated hESCs, but not from Activin A alone treated cells. Our results suggest that important tissue interactions occur in EB-based suspension culture that contribute to the complete induction of definitive endoderm and pancreas progenitors. Further differentiation occurs after EBs are embedded in Matrigel and cultured in serum-free media containing insulin, transferrin, selenium, FGF7, nicotinamide, islet neogenesis associated peptide (INGAP) and exendin-4, a long acting GLP-1 agonist. 21-28days after embedding, PDX1 gene expression levels are comparable to those of human islets used for transplantation, and many PDX1(+) clusters are formed. Almost all cells in PDX1(+) clusters co-express FOXA2, HNF1ß, HNF6 and SOX9 proteins, and many cells also express CPA1, NKX6.1 and PTF1a. If cells are then switched to medium containing B27 and nicotinamide for 7-14days, then the number of insulin(+) cells increases markedly. Our study identifies a new chemically defined culture protocol for inducing endoderm- and pancreas-committed cells from hESCs and reveals an interplay between FGF, Activin A and BMP signaling in early hESC fate determination.
Collapse
Affiliation(s)
- Xiaofang Xu
- Department of Surgery, Division of Transplantation, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin Institute of Medical Research, 600 Highland Ave., Madison, WI 53792, USA.
| | | | | |
Collapse
|
48
|
Hudson JE, Zimmermann WH. Tuning Wnt-signaling to enhance cardiomyogenesis in human embryonic and induced pluripotent stem cells. J Mol Cell Cardiol 2011; 51:277-9. [PMID: 21723872 DOI: 10.1016/j.yjmcc.2011.06.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 06/16/2011] [Accepted: 06/16/2011] [Indexed: 02/02/2023]
|
49
|
Burridge PW, Thompson S, Millrod MA, Weinberg S, Yuan X, Peters A, Mahairaki V, Koliatsos VE, Tung L, Zambidis ET. A universal system for highly efficient cardiac differentiation of human induced pluripotent stem cells that eliminates interline variability. PLoS One 2011; 6:e18293. [PMID: 21494607 PMCID: PMC3072973 DOI: 10.1371/journal.pone.0018293] [Citation(s) in RCA: 318] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 02/23/2011] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The production of cardiomyocytes from human induced pluripotent stem cells (hiPSC) holds great promise for patient-specific cardiotoxicity drug testing, disease modeling, and cardiac regeneration. However, existing protocols for the differentiation of hiPSC to the cardiac lineage are inefficient and highly variable. We describe a highly efficient system for differentiation of human embryonic stem cells (hESC) and hiPSC to the cardiac lineage. This system eliminated the variability in cardiac differentiation capacity of a variety of human pluripotent stem cells (hPSC), including hiPSC generated from CD34(+) cord blood using non-viral, non-integrating methods. METHODOLOGY/PRINCIPAL FINDINGS We systematically and rigorously optimized >45 experimental variables to develop a universal cardiac differentiation system that produced contracting human embryoid bodies (hEB) with an improved efficiency of 94.7±2.4% in an accelerated nine days from four hESC and seven hiPSC lines tested, including hiPSC derived from neonatal CD34(+) cord blood and adult fibroblasts using non-integrating episomal plasmids. This cost-effective differentiation method employed forced aggregation hEB formation in a chemically defined medium, along with staged exposure to physiological (5%) oxygen, and optimized concentrations of mesodermal morphogens BMP4 and FGF2, polyvinyl alcohol, serum, and insulin. The contracting hEB derived using these methods were composed of high percentages (64-89%) of cardiac troponin I(+) cells that displayed ultrastructural properties of functional cardiomyocytes and uniform electrophysiological profiles responsive to cardioactive drugs. CONCLUSION/SIGNIFICANCE This efficient and cost-effective universal system for cardiac differentiation of hiPSC allows a potentially unlimited production of functional cardiomyocytes suitable for application to hPSC-based drug development, cardiac disease modeling, and the future generation of clinically-safe nonviral human cardiac cells for regenerative medicine.
Collapse
Affiliation(s)
- Paul W. Burridge
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
- * E-mail: (ETZ); (PWB)
| | - Susan Thompson
- Department of Biomedical Engineering, The Johns Hopkins University School
of Medicine, Baltimore, Maryland, United States of America
| | - Michal A. Millrod
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Seth Weinberg
- Department of Biomedical Engineering, The Johns Hopkins University School
of Medicine, Baltimore, Maryland, United States of America
| | - Xuan Yuan
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Ann Peters
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Vasiliki Mahairaki
- Division of Neuropathology, Department of Pathology, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Vassilis E. Koliatsos
- Division of Neuropathology, Department of Pathology, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
- Department of Neurology, The Johns Hopkins University School of Medicine,
Baltimore, Maryland, United States of America
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
| | - Leslie Tung
- Department of Biomedical Engineering, The Johns Hopkins University School
of Medicine, Baltimore, Maryland, United States of America
| | - Elias T. Zambidis
- Johns Hopkins Institute for Cell Engineering, The Johns Hopkins
University School of Medicine, Baltimore, Maryland, United States of
America
- * E-mail: (ETZ); (PWB)
| |
Collapse
|
50
|
Cimetta E, Sirabella D, Yeager K, Davidson K, Simon J, Moon RT, Vunjak-Novakovic G. Microfluidic device generating stable concentration gradients for long term cell culture: application to Wnt3a regulation of β-catenin signaling. LAB ON A CHIP 2010; 10:3277-83. [PMID: 20936235 PMCID: PMC4106280 DOI: 10.1039/c0lc00033g] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
In developing tissues, proteins and signaling molecules present themselves in the form of concentration gradients, which determine the fate specification and behavior of the sensing cells. To mimic these conditions in vitro, we developed a microfluidic device designed to generate stable concentration gradients at low hydrodynamic shear and allowing long term culture of adhering cells. The gradient forms in a culture space between two parallel laminar flow streams of culture medium at two different concentrations of a given morphogen. The exact algorithm for defining the concentration gradients was established with the aid of mathematical modeling of flow and mass transport. Wnt3a regulation of β-catenin signaling was chosen as a case study. The highly conserved Wnt-activated β-catenin pathway plays major roles in embryonic development, stem cell proliferation and differentiation. Wnt3a stimulates the activity of β-catenin pathway, leading to translocation of β-catenin to the nucleus where it activates a series of target genes. We cultured A375 cells stably expressing a Wnt/β-catenin reporter driving the expression of Venus, pBARVS, inside the microfluidic device. The extent to which the β-catenin pathway was activated in response to a gradient of Wnt3a was assessed in real time using the BARVS reporter gene. On a single cell level, the β-catenin signaling was proportionate to the concentration gradient of Wnt3a; we thus propose that the modulation of Wnt3a gradients in real time can provide new insights into the dynamics of β-catenin pathway, under conditions that replicate some aspects of the actual cell-tissue milieu. Our device thus offers a highly controllable platform for exploring the effects of concentration gradients on cultured cells.
Collapse
Affiliation(s)
- Elisa Cimetta
- Columbia University, Department of Biomedical Engineering, Vanderbilt Clinic, VC12-234, 622 West 168th Street, New York NY 10032
- Tel: +1 (212) 305-2304; Fax: 1 (212) 305-4692;
| | - Dario Sirabella
- Columbia University, Department of Biomedical Engineering, Vanderbilt Clinic, VC12-234, 622 West 168th Street, New York NY 10032
| | - Keith Yeager
- Columbia University, Department of Biomedical Engineering, Vanderbilt Clinic, VC12-234, 622 West 168th Street, New York NY 10032
| | - Kathryn Davidson
- University of Washington School of Medicine, Institute for Stem Cell and Regenerative Medicine, HHMI and Dept. of Pharmacology, Seattle, WA
| | - Joseph Simon
- Columbia University, Department of Biomedical Engineering, Vanderbilt Clinic, VC12-234, 622 West 168th Street, New York NY 10032
| | - Randall T Moon
- University of Washington School of Medicine, Institute for Stem Cell and Regenerative Medicine, HHMI and Dept. of Pharmacology, Seattle, WA
| | - Gordana Vunjak-Novakovic
- Columbia University, Department of Biomedical Engineering, Vanderbilt Clinic, VC12-234, 622 West 168th Street, New York NY 10032
| |
Collapse
|