201
|
Exposure-based assessment of chemical teratogenicity using morphogenetic aggregates of human embryonic stem cells. Reprod Toxicol 2019; 91:74-91. [PMID: 31711903 PMCID: PMC6980740 DOI: 10.1016/j.reprotox.2019.10.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 10/17/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022]
Abstract
Pluripotent stem cells recapitulate many aspects of embryogenesis in vitro. Here, we established a novel culture system to differentiate human embryonic stem cell aggregates (HESCA), and evaluated its utility for teratogenicity assessment. Culture of HESCA with modulators of developmental signals induced morphogenetic and molecular changes associated with differentiation of the paraxial mesoderm and neuroectoderm. To examine impact of teratogenic exposures on HESCA differentiation, 18 compounds were tested, for which adequate information on in vivo plasma concentrations is available. HESCA treated with each compound were examined for gross morphology and transcript levels of 15 embryogenesis regulator genes. Significant alterations in the transcript levels were observed for 94% (15/16) of the teratogenic exposures within 5-fold margin, whereas no alteration was observed for 92% (11/12) of the non-teratogenic exposures. Our study demonstrates that transcriptional changes in HESCA serve as predictive indicator of teratogenicity in a manner comparable to in vivo exposure levels.
Collapse
|
202
|
Chhabra S, Liu L, Goh R, Kong X, Warmflash A. Dissecting the dynamics of signaling events in the BMP, WNT, and NODAL cascade during self-organized fate patterning in human gastruloids. PLoS Biol 2019; 17:e3000498. [PMID: 31613879 PMCID: PMC6814242 DOI: 10.1371/journal.pbio.3000498] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 10/25/2019] [Accepted: 09/23/2019] [Indexed: 12/22/2022] Open
Abstract
During gastrulation, the pluripotent epiblast self-organizes into the 3 germ layers-endoderm, mesoderm and ectoderm, which eventually form the entire embryo. Decades of research in the mouse embryo have revealed that a signaling cascade involving the Bone Morphogenic Protein (BMP), WNT, and NODAL pathways is necessary for gastrulation. In vivo, WNT and NODAL ligands are expressed near the site of gastrulation in the posterior of the embryo, and knockout of these ligands leads to a failure to gastrulate. These data have led to the prevailing view that a signaling gradient in WNT and NODAL underlies patterning during gastrulation; however, the activities of these pathways in space and time have never been directly observed. In this study, we quantify BMP, WNT, and NODAL signaling dynamics in an in vitro model of human gastrulation. Our data suggest that BMP signaling initiates waves of WNT and NODAL signaling activity that move toward the colony center at a constant rate. Using a simple mathematical model, we show that this wave-like behavior is inconsistent with a reaction-diffusion-based Turing system, indicating that there is no stable signaling gradient of WNT/NODAL. Instead, the final signaling state is homogeneous, and spatial differences arise only from boundary effects. We further show that the durations of WNT and NODAL signaling control mesoderm differentiation, while the duration of BMP signaling controls differentiation of CDX2-positive extra-embryonic cells. The identity of these extra-embryonic cells has been controversial, and we use RNA sequencing (RNA-seq) to obtain their transcriptomes and show that they closely resemble human trophoblast cells in vivo. The domain of BMP signaling is identical to the domain of differentiation of these trophoblast-like cells; however, neither WNT nor NODAL forms a spatial pattern that maps directly to the mesodermal region, suggesting that mesoderm differentiation is controlled dynamically by the combinatorial effect of multiple signals. We synthesize our data into a mathematical model that accurately recapitulates signaling dynamics and predicts cell fate patterning upon chemical and physical perturbations. Taken together, our study shows that the dynamics of signaling events in the BMP, WNT, and NODAL cascade in the absence of a stable signaling gradient control fate patterning of human gastruloids.
Collapse
Affiliation(s)
- Sapna Chhabra
- Systems, Synthetic and Physical Biology, Rice University, Houston, Texas, United States of America
| | - Lizhong Liu
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - Ryan Goh
- Department of Mathematics, Boston University, Boston, Massachusetts, United States of America
| | - Xiangyu Kong
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - Aryeh Warmflash
- Department of Biosciences, Rice University, Houston, Texas, United States of America
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
203
|
Sagy N, Slovin S, Allalouf M, Pour M, Savyon G, Boxman J, Nachman I. Prediction and control of symmetry breaking in embryoid bodies by environment and signal integration. Development 2019; 146:dev.181917. [PMID: 31575644 DOI: 10.1242/dev.181917] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/16/2019] [Indexed: 01/03/2023]
Abstract
During early embryogenesis, mechanical constraints and localized biochemical signals co-occur around anteroposterior axis determination and symmetry breaking. Their relative roles, however, are hard to tease apart in vivo Using brachyury (Bra), a primitive streak and mesendoderm marker in mouse embryoid bodies (EBs), we studied how contact, biochemical cues and neighboring cell cues affect the positioning of a primitive streak-like locus and thus determine the anteroposterior axis. We show that a Bra-competent layer must be formed in the EB before Bra expression initiates, and that Bra onset locus position is biased by contact points of the EB with its surrounding, probably through modulation of chemical cues rather than by mechanical signaling. We can push or pull Bra onset away from contact points by introducing a separate localized Wnt signal source, or maneuver Bra onset to a few loci or to an isotropic peripheral pattern. Furthermore, we show that Foxa2-positive cells are predictive of the future location of Bra onset, demonstrating an earlier symmetry-breaking event. Our analysis of factors affecting symmetry breaking and spatial fate choice during this developmental process could prove valuable for in vitro differentiation and organoid formation.
Collapse
Affiliation(s)
- Naor Sagy
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shaked Slovin
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Maya Allalouf
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Maayan Pour
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Gaya Savyon
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Jonathan Boxman
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Iftach Nachman
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
204
|
Zhang Z, Zwick S, Loew E, Grimley JS, Ramanathan S. Mouse embryo geometry drives formation of robust signaling gradients through receptor localization. Nat Commun 2019; 10:4516. [PMID: 31586065 PMCID: PMC6778081 DOI: 10.1038/s41467-019-12533-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 09/12/2019] [Indexed: 12/21/2022] Open
Abstract
Morphogen signals are essential for cell fate specification during embryogenesis. Some receptors that sense these morphogens are known to localize to only the apical or basolateral membrane of polarized cell lines in vitro. How such localization affects morphogen sensing and patterning in the developing embryo remains unknown. Here, we show that the formation of a robust BMP signaling gradient in the early mouse embryo depends on the restricted, basolateral localization of BMP receptors. The mis-localization of receptors to the apical membrane results in ectopic BMP signaling in the mouse epiblast in vivo. With evidence from mathematical modeling, human embryonic stem cells in vitro, and mouse embryos in vivo, we find that the geometric compartmentalization of BMP receptors and ligands creates a signaling gradient that is buffered against fluctuations. Our results demonstrate the importance of receptor localization and embryo geometry in shaping morphogen signaling during embryogenesis.
Collapse
Affiliation(s)
- Zhechun Zhang
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA.
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
| | - Steven Zwick
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Ethan Loew
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Joshua S Grimley
- Allen Institute for Brain Science, Seattle, WA, 98109, USA
- Universal Cells, Seattle, WA, 98121, USA
| | - Sharad Ramanathan
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA.
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA.
| |
Collapse
|
205
|
Frontier Progress in the Establishment of Trophoblast Stem Cell and the Identification of New Cell Subtypes at the Maternal-Fetal Interface. MATERNAL-FETAL MEDICINE 2019. [DOI: 10.1097/fm9.0000000000000023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
206
|
Hemberger M, Hanna CW, Dean W. Mechanisms of early placental development in mouse and humans. Nat Rev Genet 2019; 21:27-43. [PMID: 31534202 DOI: 10.1038/s41576-019-0169-4] [Citation(s) in RCA: 275] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2019] [Indexed: 02/08/2023]
Abstract
The importance of the placenta in supporting mammalian development has long been recognized, but our knowledge of the molecular, genetic and epigenetic requirements that underpin normal placentation has remained remarkably under-appreciated. Both the in vivo mouse model and in vitro-derived murine trophoblast stem cells have been invaluable research tools for gaining insights into these aspects of placental development and function, with recent studies starting to reshape our view of how a unique epigenetic environment contributes to trophoblast differentiation and placenta formation. These advances, together with recent successes in deriving human trophoblast stem cells, open up new and exciting prospects in basic and clinical settings that will help deepen our understanding of placental development and associated disorders of pregnancy.
Collapse
Affiliation(s)
- Myriam Hemberger
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada. .,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada. .,Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada. .,Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK. .,Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| | - Courtney W Hanna
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Wendy Dean
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada. .,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada. .,Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK. .,Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| |
Collapse
|
207
|
Vianello S, Lutolf MP. Understanding the Mechanobiology of Early Mammalian Development through Bioengineered Models. Dev Cell 2019; 48:751-763. [PMID: 30913407 DOI: 10.1016/j.devcel.2019.02.024] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/13/2019] [Accepted: 02/26/2019] [Indexed: 12/21/2022]
Abstract
Research in developmental biology has been recently enriched by a multitude of in vitro models recapitulating key milestones of mammalian embryogenesis. These models obviate the challenge posed by the inaccessibility of implanted embryos, multiply experimental opportunities, and favor approaches traditionally associated with organoids and tissue engineering. Here, we provide a perspective on how these models can be applied to study the mechano-geometrical contributions to early mammalian development, which still escape direct verification in species that develop in utero. We thus outline new avenues for robust and scalable perturbation of geometry and mechanics in ways traditionally limited to non-implanting developmental models.
Collapse
Affiliation(s)
- Stefano Vianello
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; Institute of Chemical Sciences and Engineering, School of Basic Science (SB), EPFL, Lausanne, Switzerland.
| |
Collapse
|
208
|
Kime C, Kiyonari H, Ohtsuka S, Kohbayashi E, Asahi M, Yamanaka S, Takahashi M, Tomoda K. Induced 2C Expression and Implantation-Competent Blastocyst-like Cysts from Primed Pluripotent Stem Cells. Stem Cell Reports 2019; 13:485-498. [PMID: 31402336 PMCID: PMC6739768 DOI: 10.1016/j.stemcr.2019.07.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 07/09/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022] Open
Abstract
Soon after fertilization, the few totipotent cells of mammalian embryos diverge to form a structure called the blastocyst (BC). Although numerous cell types, including germ cells and extended-pluripotency stem cells, have been developed from pluripotent stem cells (PSCs) in vitro, generating functional BCs only from PSCs remains elusive. Here, we describe induced self-organizing 3D BC-like cysts (iBLCs) generated from mouse PSC culture. Resembling natural BCs, iBLCs have a blastocoel-like cavity and were formed with outer cells expressing trophectoderm lineage markers and with inner cells expressing pluripotency markers. iBLCs transplanted to pseudopregnant mice uteruses implanted, induced decidualization, and exhibited growth and development before resorption, demonstrating that iBLCs are implantation competent. iBLC precursor intermediates required the transcription factor Prdm14 and concomitantly activated the totipotency-related cleavage-stage MERVL reporter and 2C genes. Thus, our system may contribute to the understanding of molecular mechanisms underpinning totipotency, embryogenesis, and implantation.
Collapse
Affiliation(s)
- Cody Kime
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Lab of Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan.
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Satoshi Ohtsuka
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Eiko Kohbayashi
- Second Department of Internal Medicine, Osaka Medical College, Osaka 569-8686, Japan
| | - Michio Asahi
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka 569-8686, Japan
| | - Shinya Yamanaka
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Masayo Takahashi
- Lab of Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Kiichiro Tomoda
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka 569-8686, Japan.
| |
Collapse
|
209
|
Johnson M. Human in vitro fertilisation and developmental biology: a mutually influential history. Development 2019; 146:146/17/dev183145. [PMID: 31488509 DOI: 10.1242/dev.183145] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
This article describes the origins and development of in vitro fertilisation (IVF) and how it was influenced by, and influenced, basic research in developmental biology. It describes the technical and social challenges that confronted the pioneers in this field of study, and the considerable progress that has been made since those early days. It also considers how IVF has contributed, and continues to contribute, to our understanding of early human development.
Collapse
Affiliation(s)
- Martin Johnson
- Department of Anatomy, University of Cambridge, Cambridge, CB2 3DY, UK
| |
Collapse
|
210
|
Zheng Y, Xue X, Shao Y, Wang S, Esfahani SN, Li Z, Muncie JM, Lakins JN, Weaver VM, Gumucio DL, Fu J. Controlled modelling of human epiblast and amnion development using stem cells. Nature 2019; 573:421-425. [PMID: 31511693 PMCID: PMC8106232 DOI: 10.1038/s41586-019-1535-2] [Citation(s) in RCA: 301] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/06/2019] [Indexed: 11/09/2022]
Abstract
Early human embryonic development involves extensive lineage diversification, cell-fate specification and tissue patterning1. Despite its basic and clinical importance, early human embryonic development remains relatively unexplained owing to interspecies divergence2,3 and limited accessibility to human embryo samples. Here we report that human pluripotent stem cells (hPSCs) in a microfluidic device recapitulate, in a highly controllable and scalable fashion, landmarks of the development of the epiblast and amniotic ectoderm parts of the conceptus, including lumenogenesis of the epiblast and the resultant pro-amniotic cavity, formation of a bipolar embryonic sac, and specification of primordial germ cells and primitive streak cells. We further show that amniotic ectoderm-like cells function as a signalling centre to trigger the onset of gastrulation-like events in hPSCs. Given its controllability and scalability, the microfluidic model provides a powerful experimental system to advance knowledge of human embryology and reproduction. This model could assist in the rational design of differentiation protocols of hPSCs for disease modelling and cell therapy, and in high-throughput drug and toxicity screens to prevent pregnancy failure and birth defects.
Collapse
Affiliation(s)
- Yi Zheng
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yue Shao
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Sicong Wang
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | - Zida Li
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Jonathon M Muncie
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, San Francisco, CA, USA
| | - Johnathon N Lakins
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | - Valerie M Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiation Oncology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Deborah L Gumucio
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
211
|
Corsini NS, Peer AM, Moeseneder P, Roiuk M, Burkard TR, Theussl HC, Moll I, Knoblich JA. Coordinated Control of mRNA and rRNA Processing Controls Embryonic Stem Cell Pluripotency and Differentiation. Cell Stem Cell 2019; 22:543-558.e12. [PMID: 29625069 DOI: 10.1016/j.stem.2018.03.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 01/05/2018] [Accepted: 03/07/2018] [Indexed: 12/23/2022]
Abstract
Stem cell-specific transcriptional networks are well known to control pluripotency, but constitutive cellular processes such as mRNA splicing and protein synthesis can add complex layers of regulation with poorly understood effects on cell-fate decisions. Here, we show that the RNA binding protein HTATSF1 controls embryonic stem cell differentiation by regulating multiple aspects of RNA processing during ribosome biogenesis. HTATSF1, in a complex with splicing factor SF3B1, controls intron removal from ribosomal protein transcripts and regulates ribosomal RNA transcription and processing, thereby controlling 60S ribosomal abundance and protein synthesis. HTATSF1-dependent protein synthesis is essential for naive pre-implantation epiblast to transition into post-implantation epiblast, a stage with transiently low protein synthesis, and further differentiation toward neuroectoderm. Together, these results identify coordinated regulation of ribosomal RNA and protein synthesis by HTATSF1 and show that this essential mechanism controls protein synthesis during early mammalian embryogenesis.
Collapse
Affiliation(s)
- Nina S Corsini
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Angela M Peer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Paul Moeseneder
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Mykola Roiuk
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, 1030 Vienna, Austria
| | - Thomas R Burkard
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria; Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Hans-Christian Theussl
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria; Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Isabella Moll
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, 1030 Vienna, Austria
| | - Juergen A Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria.
| |
Collapse
|
212
|
Geng T, Zhang D, Jiang W. Epigenetic Regulation of Transition Among Different Pluripotent States: Concise Review. Stem Cells 2019; 37:1372-1380. [PMID: 31339608 DOI: 10.1002/stem.3064] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/12/2019] [Accepted: 07/08/2019] [Indexed: 12/11/2022]
Abstract
The extraordinary progress of pluripotent stem cell research provides a revolutionary avenue to understand mammalian early embryonic development. Besides well-established conventional mouse and human embryonic stem cells, the discoveries of naive state human stem cell, two-cell-like cell, and the newly defined "extended pluripotent" stem cell and "expanded potential" stem cell with bidirectional chimeric ability have greatly broadened the horizons of more pluripotent states recaptured and maintained in dish, infinitely approaching the totipotent blastomere state. Although all these pluripotent cell types can self-renew and have the ability to differentiate into all the three germ layers, accumulating evidence suggests that these pluripotent states display distinct epigenetic characters. More strikingly, epigenetic reprogramming, including DNA methylation, histone modification, and chromatin remodeling, is required to reset the cell fate commitment, suggesting that epigenetic mechanisms may play an active and important role in the maintenance and transition among these pluripotent states. Here, we have reviewed studies on various pluripotent states, with a highlight on the epigenetic regulation during the interconversion. Stem Cells 2019;37:1372-1380.
Collapse
Affiliation(s)
- Ting Geng
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University; Medical Research Institute, Wuhan University; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Hubei Province, People's Republic of China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, Hubei Province, People's Republic of China
| | - Wei Jiang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University; Medical Research Institute, Wuhan University; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Hubei Province, People's Republic of China
| |
Collapse
|
213
|
Soares MJ, Varberg KM, Iqbal K. Hemochorial placentation: development, function, and adaptations. Biol Reprod 2019; 99:196-211. [PMID: 29481584 DOI: 10.1093/biolre/ioy049] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/21/2018] [Indexed: 11/12/2022] Open
Abstract
Placentation is a reproductive adaptation that permits fetal growth and development within the protected confines of the female reproductive tract. Through this important role, the placenta also determines postnatal health and susceptibility to disease. The hemochorial placenta is a prominent feature in primate and rodent development. This manuscript provides an overview of the basics of hemochorial placental development and function, provides perspectives on major discoveries that have shaped placental research, and thoughts on strategies for future investigation.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas, USA and the Center for Perinatal Research, Children΄s Research Institute, Children΄s Mercy, Kansas City, Missouri, USA
| | - Kaela M Varberg
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
214
|
Bubela T, Kleiderman E, Master Z, Ogbogu U, Ravitsky V, Zarzeczny A, Knoppers BM. Canada's Assisted Human Reproduction Act: Pragmatic Reforms in Support of Research. Front Med (Lausanne) 2019; 6:157. [PMID: 31355201 PMCID: PMC6636215 DOI: 10.3389/fmed.2019.00157] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/24/2019] [Indexed: 12/20/2022] Open
Abstract
Canada's Assisted Human Reproduction Act is long overdue for Parliamentary review. We argue that the current regulation of research using human reproductive materials is not proportionate, not responsive to the uncertain threats posed to human and environmental health and safety, and is not considerate of diverse values in a democratic society. We propose tailored regulatory carve-outs for in vitro research for currently prohibited activities, such as gene editing, and for the exercise of Ministerial Discretion for access by Canadians to experimental in vivo interventions that are currently prohibited, such as mitochondrial replacement therapy. Our recommendations are bounded by constitutional constraints that recognize political and practical challenges in keeping oversight of this research under Federal jurisdiction, whether conducted in academic or private sectors. The proposed nuanced regulatory scheme should be overseen by a new national Agency, modeled on a blend of the Canadian Stem Cell Oversight Committee and Assisted Human Reproduction Canada.
Collapse
Affiliation(s)
- Tania Bubela
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Erika Kleiderman
- Centre of Genomics and Policy, Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Zubin Master
- Mayo Clinic Center for Regenerative Medicine, Rochester, MN, United States
- Biomedical Ethics Research Program, Mayo Clinic, Rochester, MN, United States
| | - Ubaka Ogbogu
- Faculties of Law, Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Vardit Ravitsky
- Department of Social and Preventive Medicine, School of Public Health, University of Montreal, Montreal, QC, Canada
| | - Amy Zarzeczny
- Johnson Shoyama Graduate School of Public Policy, University of Regina, Regina, SK, Canada
| | - Bartha Maria Knoppers
- Centre of Genomics and Policy, Department of Human Genetics, McGill University, Montreal, QC, Canada
| |
Collapse
|
215
|
Simunovic M, Metzger JJ, Etoc F, Yoney A, Ruzo A, Martyn I, Croft G, You DS, Brivanlou AH, Siggia ED. A 3D model of a human epiblast reveals BMP4-driven symmetry breaking. Nat Cell Biol 2019; 21:900-910. [PMID: 31263269 DOI: 10.1038/s41556-019-0349-7] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/30/2019] [Indexed: 01/05/2023]
Abstract
Breaking the anterior-posterior symmetry in mammals occurs at gastrulation. Much of the signalling network underlying this process has been elucidated in the mouse; however, there is no direct molecular evidence of events driving axis formation in humans. Here, we use human embryonic stem cells to generate an in vitro three-dimensional model of a human epiblast whose size, cell polarity and gene expression are similar to a day 10 human epiblast. A defined dose of BMP4 spontaneously breaks axial symmetry, and induces markers of the primitive streak and epithelial-to-mesenchymal transition. We show that WNT signalling and its inhibitor DKK1 play key roles in this process downstream of BMP4. Our work demonstrates that a model human epiblast can break axial symmetry despite the absence of asymmetry in the initial signal and of extra-embryonic tissues or maternal cues. Our three-dimensional model is an assay for the molecular events underlying human axial symmetry breaking.
Collapse
Affiliation(s)
- Mijo Simunovic
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY, USA
- Center for Studies in Physics and Biology, The Rockefeller University, New York, NY, USA
| | - Jakob J Metzger
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY, USA
- Center for Studies in Physics and Biology, The Rockefeller University, New York, NY, USA
| | - Fred Etoc
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY, USA
- Center for Studies in Physics and Biology, The Rockefeller University, New York, NY, USA
| | - Anna Yoney
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY, USA
- Center for Studies in Physics and Biology, The Rockefeller University, New York, NY, USA
| | - Albert Ruzo
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY, USA
| | - Iain Martyn
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY, USA
- Center for Studies in Physics and Biology, The Rockefeller University, New York, NY, USA
| | - Gist Croft
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY, USA
| | | | - Ali H Brivanlou
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY, USA.
| | - Eric D Siggia
- Center for Studies in Physics and Biology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
216
|
|
217
|
Shahbazi MN, Siggia ED, Zernicka-Goetz M. Self-organization of stem cells into embryos: A window on early mammalian development. Science 2019; 364:948-951. [PMID: 31171690 PMCID: PMC8300856 DOI: 10.1126/science.aax0164] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Embryonic development is orchestrated by robust and complex regulatory mechanisms acting at different scales of organization. In vivo studies are particularly challenging for mammals after implantation, owing to the small size and inaccessibility of the embryo. The generation of stem cell models of the embryo represents a powerful system with which to dissect this complexity. Control of geometry, modulation of the physical environment, and priming with chemical signals reveal the intrinsic capacity of embryonic stem cells to make patterns. Adding the stem cells for the extraembryonic lineages generates three-dimensional models that are more autonomous from the environment and recapitulate many features of the pre- and postimplantation mouse embryo, including gastrulation. Here, we review the principles of self-organization and how they set cells in motion to create an embryo.
Collapse
Affiliation(s)
- Marta N Shahbazi
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK.
| | - Eric D Siggia
- Center for Studies in Physics and Biology, The Rockefeller University, New York, NY 10065, USA.
| | - Magdalena Zernicka-Goetz
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK.
| |
Collapse
|
218
|
Abstract
Organoids are multicellular structures that can be derived from adult organs or pluripotent stem cells. Early versions of organoids range from simple epithelial structures to complex, disorganized tissues with large cellular diversity. The current challenge is to engineer cellular complexity into organoids in a controlled manner that results in organized assembly and acquisition of tissue function. These efforts have relied on studies of organ assembly during embryonic development and have resulted in the development of organoids with multilayer tissue complexity and higher-order functions. We discuss how the next generation of organoids can be designed by means of an engineering-based narrative design to control patterning, assembly, morphogenesis, growth, and function.
Collapse
Affiliation(s)
- Takanori Takebe
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Institute of Research, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - James M Wells
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
219
|
McMahon R, Sibbritt T, Salehin N, Osteil P, Tam PPL. Mechanistic insights from the LHX1-driven molecular network in building the embryonic head. Dev Growth Differ 2019; 61:327-336. [PMID: 31111476 DOI: 10.1111/dgd.12609] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/05/2019] [Accepted: 04/08/2019] [Indexed: 12/27/2022]
Abstract
Development of an embryo is driven by a series of molecular instructions that control the differentiation of tissue precursor cells and shape the tissues into major body parts. LIM homeobox 1 (LHX1) is a transcription factor that plays a major role in the development of the embryonic head of the mouse. Loss of LHX1 function disrupts the morphogenetic movement of head tissue precursors and impacts on the function of molecular factors in modulating the activity of the WNT signaling pathway. LHX1 acts with a transcription factor complex to regulate the transcription of target genes in multiple phases of development and in a range of embryonic tissues of the mouse and Xenopus. Determining the interacting factors and transcriptional targets of LHX1 will be key to unraveling the ensemble of factors involved in head development and building a head gene regulatory network.
Collapse
Affiliation(s)
- Riley McMahon
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Westmead, New South Wales, Australia
| | - Tennille Sibbritt
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Westmead, New South Wales, Australia
| | - Nazmus Salehin
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Westmead, New South Wales, Australia
| | - Pierre Osteil
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Westmead, New South Wales, Australia
| | - Patrick P L Tam
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Westmead, New South Wales, Australia
| |
Collapse
|
220
|
Ebrahimkhani MR, Ebisuya M. Synthetic developmental biology: build and control multicellular systems. Curr Opin Chem Biol 2019; 52:9-15. [PMID: 31102790 DOI: 10.1016/j.cbpa.2019.04.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 02/08/2023]
Abstract
Synthetic biology offers a bottom-up engineering approach that intends to understand complex systems via design-build-test cycles. Embryonic development comprises complex processes that originate at the level of gene regulatory networks in a cell and emerge into collective cellular behaviors with multicellular forms and functions. Here, we review synthetic biology approaches to development that involve building de novo developmental trajectories or engineering control in stem cell-derived multicellular systems. The field of synthetic developmental biology is rapidly growing with the help of recent advances in artificial gene circuits, self-organizing organoids, and controllable tissue microenvironments. The outcome will be a blueprint to decode principles of morphogenesis and to create programmable organoids with novel designs or improved functions.
Collapse
Affiliation(s)
- Mo R Ebrahimkhani
- Biodesign Institute, Arizona State Tempe, AZ, USA; School of Biological and Health Systems Engineering, Arizona State Tempe, AZ, USA; Mayo Clinic College of Medicine and Science, Phoenix, AZ, USA.
| | - Miki Ebisuya
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader, 88, 08003, Barcelona, Spain.
| |
Collapse
|
221
|
Ma S. Microfluidics tubing as a synthesizer for ordered microgel networks. SOFT MATTER 2019; 15:3848-3853. [PMID: 31032832 DOI: 10.1039/c9sm00626e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Ordered microgel networks have undergone extensive research and shown translational promise in tissue engineering, precision and regenerative medicine, controlled delivery, optics and electronics, etc. Here, we introduce a new low-cost and efficient synthesizer for ordered microgel networks. The gel precursor microdroplets are formulated and incubated in a microfluidics tubing system to obtain tailorable and reproducible microgels, which are then patterned into networks under the precise spatiotemporal control of the tubing system and integrated either by crosslinking the microgel interfaces or by forming lipid bilayers at the interfaces. The system can synthesize ordered networks out of heterogeneous microgels by withdrawing multi-phase cell-laden or acellular gel precursors into the tubing and gelation, or out of homogeneous microgels by simultaneously injecting gel precursors and immiscible oil into the tubing and gelation. The ordered gel networks are synthesized at the tubing outlet or within a piece of enlarging tubing, where the microgels are collided and glued in defined sequences.
Collapse
Affiliation(s)
- Shaohua Ma
- Shenzhen Engineering Laboratory for Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen 518055, P. R. China.
| |
Collapse
|
222
|
Sybirna A, Wong FCK, Surani MA. Genetic basis for primordial germ cells specification in mouse and human: Conserved and divergent roles of PRDM and SOX transcription factors. Curr Top Dev Biol 2019; 135:35-89. [PMID: 31155363 DOI: 10.1016/bs.ctdb.2019.04.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Primordial germ cells (PGCs) are embryonic precursors of sperm and egg that pass on genetic and epigenetic information from one generation to the next. In mammals, they are induced from a subset of cells in peri-implantation epiblast by BMP signaling from the surrounding tissues. PGCs then initiate a unique developmental program that involves comprehensive epigenetic resetting and repression of somatic genes. This is orchestrated by a set of signaling molecules and transcription factors that promote germ cell identity. Here we review significant findings on mammalian PGC biology, in particular, the genetic basis for PGC specification in mice and human, which has revealed an evolutionary divergence between the two species. We discuss the importance and potential basis for these differences and focus on several examples to illustrate the conserved and divergent roles of critical transcription factors in mouse and human germline.
Collapse
Affiliation(s)
- Anastasiya Sybirna
- Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom; Physiology, Development and Neuroscience Department, University of Cambridge, Cambridge, United Kingdom; Wellcome Trust/Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.
| | - Frederick C K Wong
- Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom; Physiology, Development and Neuroscience Department, University of Cambridge, Cambridge, United Kingdom
| | - M Azim Surani
- Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom; Physiology, Development and Neuroscience Department, University of Cambridge, Cambridge, United Kingdom; Wellcome Trust/Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
223
|
Regier MC, Tokar JJ, Warrick JW, Pabon L, Berthier E, Beebe DJ, Stevens KR. User-defined morphogen patterning for directing human cell fate stratification. Sci Rep 2019; 9:6433. [PMID: 31015521 PMCID: PMC6478938 DOI: 10.1038/s41598-019-42874-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023] Open
Abstract
Concentration gradients of biochemical stimuli such as morphogens play a critical role in directing cell fate patterning across species and throughout development but are not commonly recapitulated in vitro. While in vitro biomolecule gradients have been generated using customized microfluidic platforms, broad implementation has been limited because these platforms introduce new variables to cell culture such as externally driven flow, culture in a specialized matrix, or extended time for in situ long range diffusion. Here we introduce a method that enables preforming and then transferring user-controlled gradients to cells in standard "open" cultures. Our gradient patterning devices are modular and decoupled from the culture substrate. We find that gradient generation and transfer are predictable by finite element modeling and that device and loading parameters can be used to tune the stimulus pattern. Furthermore, we demonstrate use of these devices to spatially define morphogen signal gradients and direct peri-gastrulation fate stratification of human pluripotent stem cells. This method for extrinsic application of biochemical signal gradients can thus be used to spatially influence cellular fate decisions in a user-controlled manner.
Collapse
Affiliation(s)
- Mary C Regier
- Department of Bioengineering, University of Washington, 98195, Seattle, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 98109, Seattle, USA
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
| | - Jacob J Tokar
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
| | - Jay W Warrick
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
- McArdle Laboratory for Cancer Research, University of Wisconsin - Madison, 53705, Madison, USA
| | - Lil Pabon
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 98109, Seattle, USA
- Department of Pathology, University of Washington, 98195, Seattle, USA
| | - Erwin Berthier
- Department of Chemistry, University of Washington, 98195, Seattle, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
| | - Kelly R Stevens
- Department of Bioengineering, University of Washington, 98195, Seattle, USA.
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 98109, Seattle, USA.
- Department of Pathology, University of Washington, 98195, Seattle, USA.
| |
Collapse
|
224
|
In vitro generation of mouse polarized embryo-like structures from embryonic and trophoblast stem cells. Nat Protoc 2019; 13:1586-1602. [PMID: 29988106 DOI: 10.1038/s41596-018-0005-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mammalian embryogenesis requires the coordination of embryonic and extra-embryonic tissues to enable implantation into the uterus and post-implantation development to establish the body plan. Mouse embryonic stem cells (ESCs) are a useful tool for studying pluripotent embryonic tissue in vitro. However, they cannot undertake correct embryogenesis alone. Many attempts to model the early embryo in vitro involve the aggregation of ESCs into spheroids of variable size and cell number that undertake germ-layer specification but fail to recapitulate the characteristic architecture and arrangement of tissues of the early embryo. Here, we describe a protocol to generate the first embryo-like structures by directing the assembly of mouse ESCs and extra-embryonic trophoblast stem cells (TSCs) in a 3D extracellular matrix (ECM) into structures we call 'polarized embryo-like structures'. By establishing the medium and culture conditions needed to support the growth of both stem cell types simultaneously, embryonic architecture is generated within 4 d of co-culture. This protocol can be performed by those proficient in standard ESC culture techniques and can be used in developmental studies to investigate the interactions between embryonic and extra-embryonic tissues during mammalian development.
Collapse
|
225
|
Transcriptomic analysis of the interaction of choriocarcinoma spheroids with receptive vs. non-receptive endometrial epithelium cell lines: an in vitro model for human implantation. J Assist Reprod Genet 2019; 36:857-873. [PMID: 30972518 DOI: 10.1007/s10815-019-01442-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/19/2019] [Indexed: 10/27/2022] Open
Abstract
PURPOSE Several in vitro systems have been reported to model human implantation; however, the molecular dynamics of the trophoblast vs. the epithelial substrate during attachment have not been described. We have established an in vitro model which allowed us to dissect the transcriptional responses of the trophoblast and the receptive vs. non-receptive epithelium after co-culture. METHODS We established an in vitro system based on co-culture of (a) immortalized cells representing receptive (Ishikawa) or non-receptive (HEC-1-A) endometrial epithelium with (b) spheroids of a trophoblastic cell line (JEG-3) modified to express green fluorescent protein (GFP). After 48 h of co-culture, GFP+ (trophoblast cells) and GFP- cell fractions (receptive or non-receptive epithelial cells) were isolated by fluorescence-activated flow cytometry (FACS) and subjected to RNA-seq profiling and gene set enrichment analysis (GSEA). RESULTS Compared to HEC-1-A, the trophoblast challenge to Ishikawa cells differentially regulated the expression of 495 genes, which mainly involved cell adhesion and extracellular matrix (ECM) molecules. GSEA revealed enrichment of pathways related to cell division, cell cycle regulation, and metabolism in the Ishikawa substrate. Comparing the gene expression profile of trophoblast spheroids revealed that 1877 and 323 genes were upregulated or downregulated when co-cultured on Ishikawa substrates (compared to HEC-1-A), respectively. Pathways favorable to development, including tissue remodeling, organogenesis, and angiogenesis, were enhanced in the trophoblast compartment after co-culture of spheroids with receptive epithelium. By contrast, the co-culture with less receptive epithelium enriched pathways mainly related to trophoblast cell proliferation and cell cycle regulation. CONCLUSIONS Endometrial receptivity requires a transcriptional signature that determines the trophoblast response and drives attachment.
Collapse
|
226
|
Sharma S, Wistuba J, Pock T, Schlatt S, Neuhaus N. Spermatogonial stem cells: updates from specification to clinical relevance. Hum Reprod Update 2019; 25:275-297. [DOI: 10.1093/humupd/dmz006] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 11/23/2018] [Accepted: 02/22/2019] [Indexed: 12/20/2022] Open
Affiliation(s)
- Swati Sharma
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer Campus 1, Building D11, Münster, Germany
| | - Joachim Wistuba
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer Campus 1, Building D11, Münster, Germany
| | - Tim Pock
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer Campus 1, Building D11, Münster, Germany
| | - Stefan Schlatt
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer Campus 1, Building D11, Münster, Germany
| | - Nina Neuhaus
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Albert-Schweitzer Campus 1, Building D11, Münster, Germany
| |
Collapse
|
227
|
|
228
|
Samal P, van Blitterswijk C, Truckenmüller R, Giselbrecht S. Grow with the Flow: When Morphogenesis Meets Microfluidics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805764. [PMID: 30767289 DOI: 10.1002/adma.201805764] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/04/2019] [Indexed: 06/09/2023]
Abstract
Developmental biology has advanced the understanding of the intricate and dynamic processes involved in the formation of an organism from a single cell. However, many gaps remain in the knowledge of embryonic development, especially regarding tissue morphogenesis. A possible approach to mimic such phenomena uses pluripotent stem cells in in vitro morphogenetic models. Herein, these systems are summarized with emphasis on the ability to better manipulate and control cellular interfaces with either liquid or solid materials using microengineered tools, which is critical for attaining deeper insights into pattern formation and stem cell differentiation during organogenesis. The role of conventional and customized cell-culture systems in supporting important advances in the field of morphogenesis is discussed, and the fascinating role that material sciences and microengineering currently play and are expected to play in the future is highlighted. In conclusion, it is proffered that continued microfluidics innovations when applied to morphogenesis promise to provide important insights to advance many multidisciplinary fields, including regenerative medicine.
Collapse
Affiliation(s)
- Pinak Samal
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Clemens van Blitterswijk
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Roman Truckenmüller
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Stefan Giselbrecht
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| |
Collapse
|
229
|
Barciszewski J, Ciemerych MA, Twardowski T. Novel insights and innovations in biotechnology towards improved quality of life. N Biotechnol 2019; 49:58-65. [PMID: 30194997 DOI: 10.1016/j.nbt.2018.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 09/03/2018] [Accepted: 09/03/2018] [Indexed: 12/26/2022]
Abstract
Resolution of old problems with new tools seems to be a new way of challenging and finding the keys to innovation. A holistic view of different branches of science and industry, including services for society, is critically important for the development of modern science. The International Congress Eurobiotech 2017 was a special opportunity for such a universal view. In this paper, we discuss the application of different small RNAs, stem cells, epigenetics and sequencing, as well as art and bioeconomy. Our most significant message is not a new one but is very universal: biotechnology is vital for society.
Collapse
Affiliation(s)
- Jan Barciszewski
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61-704 Poznan, Poland
| | - Maria A Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Tomasz Twardowski
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61-704 Poznan, Poland.
| |
Collapse
|
230
|
Hurlbut JB, Hyun I, Levine AD, Lovell-Badge R, Lunshof JE, Matthews KRW, Mills P, Murdoch A, Pera MF, Scott CT, Tizzard J, Warnock M, Zernicka-Goetz M, Zhou Q, Zoloth L. Revisiting the Warnock rule. Nat Biotechnol 2019; 35:1029-1042. [PMID: 29121021 DOI: 10.1038/nbt.4015] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- J Benjamin Hurlbut
- School of Life Sciences, Center for Biology and Society and School for the Future of Innovation in Society, Arizona State University, Tempe, Arizona, USA
| | - Insoo Hyun
- Department of Bioethics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Aaron D Levine
- School of Public Policy, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | | | - Jeantine E Lunshof
- Department of Genetics, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands, and Department of Genetics, Church laboratory, Harvard Medical School, Boston, Massachusetts, USA
| | - Kirstin R W Matthews
- Baker Institute Center for Health and Biosciences, Rice University, Houston, TX, USA
| | | | - Alison Murdoch
- Department of Reproductive Medicine, International Centre for Life, Newcastle upon Tyne, UK
| | | | | | - Juliet Tizzard
- The Human Fertilisation and Embryology Authority, London, UK
| | - Mary Warnock
- Honorary Fellow, Hertford College, University of Oxford, Catte Street, Oxford, UK
| | | | - Qi Zhou
- The State Key Laboratory of Stem cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Laurie Zoloth
- University of Chicago Divinity School, Chicago, Illinois, USA
| |
Collapse
|
231
|
Ando Y, Okeyo KO, Adachi T. Modulation of adhesion microenvironment using mesh substrates triggers self-organization and primordial germ cell-like differentiation in mouse ES cells. APL Bioeng 2019; 3:016102. [PMID: 31069335 PMCID: PMC6481735 DOI: 10.1063/1.5072761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 03/07/2019] [Indexed: 12/21/2022] Open
Abstract
The cell adhesion microenvironment plays contributory roles in the induction of self-organized tissue formation and differentiation of pluripotent stem cells (PSCs). However, physical factors emanating from the adhesion microenvironment have been less investigated largely in part due to overreliance on biochemical approaches utilizing cytokines to drive in vitro developmental processes. Here, we report that a mesh culture technique can potentially induce mouse embryonic stem cells (mESCs) to self-organize and differentiate into cells expressing key signatures of primordial germ cells (PGCs) even with pluripotency maintained in the culture medium. Intriguingly, mESCs cultured on mesh substrates consisting of thin (5 μm-wide) strands and considerably large (200 μm-wide) openings which were set suspended in order to minimize the cell-substrate adhesion area, self-organized into cell sheets relying solely on cell-cell interactions to fill the large mesh openings by Day 2, and further into dome-shaped features around Day 6. Characterization using microarray analysis and immunofluorescence microscopy revealed that sheet-forming cells exhibited differential gene expressions related to PGCs as early as Day 2, but not other lineages such as epiblast, primitive endoderm, and trophectoderm, implying that the initial interaction with the mesh microenvironment and subsequent self-organization into cells sheets might have triggered PGC-like differentiation to occur differently from the previously reported pathway via epiblast-like differentiation. Overall, considering that the observed differentiation occurred without addition of known biochemical inducers, this study highlights that bioengineering techniques for modulating the adhesion microenvironment alone can be harnessed to coax PSCs to self-organize and differentiate, in this case, to a PGC-like state.
Collapse
|
232
|
Taniguchi K, Heemskerk I, Gumucio DL. Opening the black box: Stem cell-based modeling of human post-implantation development. J Cell Biol 2019; 218:410-421. [PMID: 30552099 PMCID: PMC6363460 DOI: 10.1083/jcb.201810084] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/27/2018] [Accepted: 12/03/2018] [Indexed: 01/06/2023] Open
Abstract
Proper development of the human embryo following its implantation into the uterine wall is critical for the successful continuation of pregnancy. However, the complex cellular and molecular changes that occur during this post-implantation period of human development are not amenable to study in vivo. Recently, several new embryo-like human pluripotent stem cell (hPSC)-based platforms have emerged, which are beginning to illuminate the current black box state of early human post-implantation biology. In this review, we will discuss how these experimental models are carving a way for understanding novel molecular and cellular mechanisms during early human development.
Collapse
Affiliation(s)
- Kenichiro Taniguchi
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Idse Heemskerk
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Deborah L Gumucio
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
233
|
Zhang S, Chen T, Chen N, Gao D, Shi B, Kong S, West RC, Yuan Y, Zhi M, Wei Q, Xiang J, Mu H, Yue L, Lei X, Wang X, Zhong L, Liang H, Cao S, Belmonte JCI, Wang H, Han J. Implantation initiation of self-assembled embryo-like structures generated using three types of mouse blastocyst-derived stem cells. Nat Commun 2019; 10:496. [PMID: 30700702 PMCID: PMC6353907 DOI: 10.1038/s41467-019-08378-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/04/2019] [Indexed: 11/09/2022] Open
Abstract
Spatially ordered embryo-like structures self-assembled from blastocyst-derived stem cells can be generated to mimic embryogenesis in vitro. However, the assembly system and developmental potential of such structures needs to be further studied. Here, we devise a nonadherent-suspension-shaking system to generate self-assembled embryo-like structures (ETX-embryoids) using mouse embryonic, trophoblast and extra-embryonic endoderm stem cells. When cultured together, the three cell types aggregate and sort into lineage-specific compartments. Signaling among these compartments results in molecular and morphogenic events that closely mimic those observed in wild-type embryos. These ETX-embryoids exhibit lumenogenesis, asymmetric patterns of gene expression for markers of mesoderm and primordial germ cell precursors, and formation of anterior visceral endoderm-like tissues. After transplantation into the pseudopregnant mouse uterus, ETX-embryoids efficiently initiate implantation and trigger the formation of decidual tissues. The ability of the three cell types to self-assemble into an embryo-like structure in vitro provides a powerful model system for studying embryogenesis.
Collapse
Affiliation(s)
- Shaopeng Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Tianzhi Chen
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Naixin Chen
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Dengfeng Gao
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Bingbo Shi
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Shuangbo Kong
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, Fujian, 361102, China
| | | | - Ye Yuan
- Colorado Center for Reproductive Medicine, Lone Tree, CO, 80124, USA
| | - Minglei Zhi
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Qingqing Wei
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Jinzhu Xiang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Haiyuan Mu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Liang Yue
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Xiaohua Lei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuepeng Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Liang Zhong
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hui Liang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Suying Cao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | | | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, Fujian, 361102, China
| | - Jianyong Han
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China. .,Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
234
|
Guo YL. The underdeveloped innate immunity in embryonic stem cells: The molecular basis and biological perspectives from early embryogenesis. Am J Reprod Immunol 2019; 81:e13089. [PMID: 30614149 DOI: 10.1111/aji.13089] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 12/31/2018] [Accepted: 01/01/2019] [Indexed: 12/21/2022] Open
Abstract
Embryonic stem cells (ESCs) have been intensively studied as a promising cell source for regenerative medicine. The rapid advancements in the field have not only proven the feasibility of ESC-based cell therapy, but also led to a better understanding of pluripotent stem cells (PSCs) as a unique cell population at an early stage of embryogenesis. Recent studies have revealed that both human and mouse ESCs have attenuated innate immune responses to infectious agents and inflammatory cytokines. These findings raise interesting questions about the rationale for ESCs, the PSCs experimentally derived from preimplantation stage embryos, to not have an innate defense mechanism that has been adapted so well in somatic cells. All somatic cells have innate immune systems that can be activated by pathogen-associated molecular patterns (PAMPs) or cellular damage-associated molecular patterns (DAMPs), leading to production of cytokines. The underdeveloped innate immunity represents a unique property of PSCs that may have important implications. This review discusses the immunological properties of PSCs, the molecular basis underlying their diminished innate immune responses, and the hypothesis that the attenuated innate immune responses could be an adaptive mechanism that allows PSCs to avoid cytotoxicity associated with inflammation and immune responses during early embryogenesis.
Collapse
Affiliation(s)
- Yan-Lin Guo
- Department of Cell and Molecular Biology, University of Southern Mississippi, Hattiesburg, Mississippi
| |
Collapse
|
235
|
Pennings S, Revuelta A, McLaughlin KA, Abd Hadi NA, Petchreing P, Ottaviano R, Meehan RR. Dynamics and Mechanisms of DNA Methylation Reprogramming. EPIGENETICS AND REGENERATION 2019:19-45. [DOI: 10.1016/b978-0-12-814879-2.00002-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
236
|
|
237
|
Ogbogu U, Zarzeczny A, Baltz J, Bedford P, Du J, Hyun I, Jaafar Y, Jurisicova A, Kleiderman E, Koukio Y, Knoppers BM, Leader A, Master Z, Nguyen MT, Noohi F, Ravitsky V, Toews M. Research on Human Embryos and Reproductive Materials: Revisiting Canadian Law and Policy. ACTA ACUST UNITED AC 2018; 13:10-19. [PMID: 29595433 PMCID: PMC5863866 DOI: 10.12927/hcpol.2018.25401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Research involving human embryos and reproductive materials, including certain forms of stem cell and genetic research, is a fast-moving area of science with demonstrated clinical relevance. Canada's current governance framework for this field of research urgently requires review and reconsideration in view of emerging applications. Based on a workshop involving ethics, legal, policy, scientific and clinical experts, we present a series of recommendations with the goal of informing and supporting health policy and decision-making regarding the governance of the field. With a pragmatic and principled governance approach, Canada can continue its global leadership in this field, as well as advance the long-term health and well-being of Canadians.
Collapse
Affiliation(s)
- Ubaka Ogbogu
- Assistant Professor, Faculties of Law and Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, AB
| | - Amy Zarzeczny
- Associate Professor, Johnson-Shoyama Graduate School of Public Policy, University of Regina, Regina, SK
| | - Jay Baltz
- Senior Scientist and Professor, Ottawa Hospital Research Institute, Faculty of Medicine, University of Ottawa, Ottawa, ON
| | - Patrick Bedford
- Manager of Clinical Translation and Regulatory Affairs, Centre for Commercialization of Regenerative Medicine (CCRM), Toronto, ON
| | - Jenny Du
- Research Trainee, Faculty of Law, University of Alberta, Edmonton, AB
| | - Insoo Hyun
- Associate Professor, Department of Bioethics, Case Western Reserve University, Cleveland, OH
| | - Yasmeen Jaafar
- Research Trainee, Faculty of Law, University of Alberta, Edmonton, AB
| | - Andrea Jurisicova
- Associate Professor, Department of Obstetrics and Gynecology, University of Toronto, Investigator, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON
| | - Erika Kleiderman
- Academic Associate, Centre of Genomics and Policy, Faculty of Medicine, Human Genetics, McGill University, Montreal, QC
| | | | - Bartha Maria Knoppers
- Professor and Director, Centre of Genomics and Policy, Faculty of Medicine, Human Genetics, McGill University, Montreal, QC
| | - Arthur Leader
- Professor of Reproductive Medicine, Department of Obstetrics and Gynecology, University of Ottawa, Ottawa, ON
| | - Zubin Master
- Associate Consultant II, Biomedical Ethics Research Program, Mayo Clinic, Rochester, MN
| | - Minh Thu Nguyen
- Academic Associate, Centre of Genomics and Policy, Faculty of Medicine, Human Genetics, McGill University, Montreal, QC
| | - Forough Noohi
- PhD Candidate, Centre of Genomics and Policy, Faculty of Medicine, Human Genetics, McGill University, Montreal, QC
| | - Vardit Ravitsky
- Associate Professor, Bioethics Programs, School of Public Health, University of Montreal, Montreal, QC
| | - Maeghan Toews
- Lecturer, Faculty of Law, University of Adelaide, Adelaide, AU
| |
Collapse
|
238
|
Beccari L, Moris N, Girgin M, Turner DA, Baillie-Johnson P, Cossy AC, Lutolf MP, Duboule D, Arias AM. Multi-axial self-organization properties of mouse embryonic stem cells into gastruloids. Nature 2018; 562:272-276. [PMID: 30283134 DOI: 10.1038/s41586-018-0578-0] [Citation(s) in RCA: 289] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 08/13/2018] [Indexed: 12/11/2022]
Abstract
The emergence of multiple axes is an essential element in the establishment of the mammalian body plan. This process takes place shortly after implantation of the embryo within the uterus and relies on the activity of gene regulatory networks that coordinate transcription in space and time. Whereas genetic approaches have revealed important aspects of these processes1, a mechanistic understanding is hampered by the poor experimental accessibility of early post-implantation stages. Here we show that small aggregates of mouse embryonic stem cells (ESCs), when stimulated to undergo gastrulation-like events and elongation in vitro, can organize a post-occipital pattern of neural, mesodermal and endodermal derivatives that mimic embryonic spatial and temporal gene expression. The establishment of the three major body axes in these 'gastruloids'2,3 suggests that the mechanisms involved are interdependent. Specifically, gastruloids display the hallmarks of axial gene regulatory systems as exemplified by the implementation of collinear Hox transcriptional patterns along an extending antero-posterior axis. These results reveal an unanticipated self-organizing capacity of aggregated ESCs and suggest that gastruloids could be used as a complementary system to study early developmental events in the mammalian embryo.
Collapse
Affiliation(s)
- Leonardo Beccari
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
| | - Naomi Moris
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Mehmet Girgin
- School of Life Sciences, Federal Institute of Technology EPFL, Lausanne, Switzerland
| | - David A Turner
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Peter Baillie-Johnson
- Department of Genetics, University of Cambridge, Cambridge, UK
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Anne-Catherine Cossy
- Swiss Cancer Research Institute (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Matthias P Lutolf
- School of Life Sciences, Federal Institute of Technology EPFL, Lausanne, Switzerland
| | - Denis Duboule
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.
- Swiss Cancer Research Institute (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | | |
Collapse
|
239
|
Metzger JJ, Simunovic M, Brivanlou AH. Synthetic embryology: controlling geometry to model early mammalian development. Curr Opin Genet Dev 2018; 52:86-91. [PMID: 29957587 PMCID: PMC6911727 DOI: 10.1016/j.gde.2018.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/30/2018] [Accepted: 06/04/2018] [Indexed: 12/15/2022]
Abstract
Differentiation of embryonic stem cells in vitro is an important tool in dissecting and understanding the mechanisms that govern early embryologic development. In recent years, there has been considerable progress in creating organoids that model gastrulation, neurulation or organogenesis. However, one of the key challenges is reproducibility. Geometrically confining stem cell colonies considerably improves reproducibility and provides quantitative control over differentiation and tissue shape. Here, we review recent advances in controlling the two-dimensional or three-dimensional organization of cells and the effect on differentiation phenotypes. Improved methods of geometrical control will allow for an even more detailed understanding of the mechanisms underlying embryologic development and will eventually pave the way for the highly reproducible generation of specific tissue types.
Collapse
Affiliation(s)
- Jakob J Metzger
- Center for Studies in Physics and Biology, The Rockefeller University, USA; Laboratory for Stem Cell Biology and Molecular Embryology, The Rockefeller University, USA
| | - Mijo Simunovic
- Center for Studies in Physics and Biology, The Rockefeller University, USA; Laboratory for Stem Cell Biology and Molecular Embryology, The Rockefeller University, USA
| | - Ali H Brivanlou
- Laboratory for Stem Cell Biology and Molecular Embryology, The Rockefeller University, USA.
| |
Collapse
|
240
|
Plusa B, Hadjantonakis AK. (De)constructing the blastocyst: Lessons in self-organization from the mouse. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.coisb.2018.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
241
|
Blin G, Wisniewski D, Picart C, Thery M, Puceat M, Lowell S. Geometrical confinement controls the asymmetric patterning of brachyury in cultures of pluripotent cells. Development 2018; 145:dev166025. [PMID: 30115626 PMCID: PMC6176930 DOI: 10.1242/dev.166025] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 07/30/2018] [Indexed: 01/02/2023]
Abstract
Diffusible signals are known to orchestrate patterning during embryogenesis, yet diffusion is sensitive to noise. The fact that embryogenesis is remarkably robust suggests that additional layers of regulation reinforce patterning. Here, we demonstrate that geometrical confinement orchestrates the spatial organisation of initially randomly positioned subpopulations of spontaneously differentiating mouse embryonic stem cells. We use micropatterning in combination with pharmacological manipulations and quantitative imaging to dissociate the multiple effects of geometry. We show that the positioning of a pre-streak-like population marked by brachyury (T) is decoupled from the size of its population, and that breaking radial symmetry of patterns imposes polarised patterning. We provide evidence for a model in which the overall level of diffusible signals together with the history of the cell culture define the number of T+ cells, whereas geometrical constraints guide patterning in a multi-step process involving a differential response of the cells to multicellular spatial organisation. Our work provides a framework for investigating robustness of patterning and provides insights into how to guide symmetry-breaking events in aggregates of pluripotent cells.
Collapse
Affiliation(s)
- Guillaume Blin
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Darren Wisniewski
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Catherine Picart
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Manuel Thery
- Univ. Grenoble-Alpes, CEA, CNRS, INRA, Biosciences and Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire and Végétale, UMR5168, CytoMorpho Lab, 38054 Grenoble, France
- Univ. Paris Diderot, CEA, INSERM, Hôpital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, CytoMorpho Lab, 75010 Paris, France
| | - Michel Puceat
- INSERM U1251, Université Aix-Marseille, MMG, 13885 Marseille, France
| | - Sally Lowell
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, EH16 4UU, UK
| |
Collapse
|
242
|
|
243
|
Abstract
Birth defects are a common occurrence in the United States and worldwide. Currently, evaluation of potential developmental toxicants (i.e., teratogens) relies heavily on animal-based models which do not always adequately mimic human development. In order to address this, researchers are developing in vitro human-based models which utilize human pluripotent stem cells (hPSCs) to assess the teratogenic potential of chemical substances. The field of human developmental toxicity assays includes a variety of platforms including monolayer, micropattern, embryoid body, and 3D organoid cultures. In this review, we will overview the field of human teratogenic assays, detail the most recent advances, and discuss current limitations and future perspectives.
Collapse
Affiliation(s)
- Kathryn E Worley
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Biotech 2147, 110 8th Street, Troy, NY 12180, USA.
| | | | | | | |
Collapse
|
244
|
Mattawanon N, Spencer JB, Schirmer DA, Tangpricha V. Fertility preservation options in transgender people: A review. Rev Endocr Metab Disord 2018; 19:231-242. [PMID: 30219984 DOI: 10.1007/s11154-018-9462-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Gender affirming procedures adversely affect the reproductive potential of transgender people. Thus, fertility preservation options should be discussed with all transpeople before medical and surgical transition. In transwomen, semen cryopreservation is typically straightforward and widely available at fertility centers. The optimal number of vials frozen depends on their reproductive goals and treatment options, therefore a consultation with a fertility specialist is optimal. Experimental techniques including spermatogonium stem cells (SSC) and testicular tissue preservation are technologies currently under development in prepubertal individuals but are not yet clinically available. In transmen, embryo and/or oocyte cryopreservation is currently the best option for fertility preservation. Embryo cryopreservation requires fertilization of the transman's oocytes with a donor or partner's sperm prior to cryopreservation, but this limits his future options for fertilizing the eggs with another partner or donor. Oocyte cryopreservation offers transmen the opportunity to preserve their fertility without committing to a male partner or sperm donor at the time of cryopreservation. Both techniques however require at least a two-week treatment course, egg retrieval under sedation and considerable cost. Ovarian tissue cryopreservation is a promising experimental method that may be performed at the same time as gender affirming surgery but is offered in only a limited amount of centers worldwide. In select places, this method may be considered for prepubertal children, adolescents, and adults when ovarian stimulation is not possible. Novel methods such as in-vitro activation of primordial follicles, in vitro maturation of immature oocytes and artificial gametes are under development and may hold promise for the future.
Collapse
Affiliation(s)
- Natnita Mattawanon
- Division of Reproductive Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Jessica B Spencer
- Division of Reproductive Endocrinology, Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, USA
| | - David A Schirmer
- Division of Reproductive Endocrinology, Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Vin Tangpricha
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| |
Collapse
|
245
|
Abstract
We present an overview of symmetry breaking in early mammalian development as a continuous process from compaction to specification of the body axes. While earlier studies have focused on individual symmetry-breaking events, recent advances enable us to explore progressive symmetry breaking during early mammalian development. Although we primarily discuss embryonic development of the mouse, as it is the best-studied mammalian model system to date, we also highlight the shared and distinct aspects between different mammalian species. Finally, we discuss how insights gained from studying mammalian development can be generalized in light of self-organization principles. With this review, we hope to highlight new perspectives in studying symmetry breaking and self-organization in multicellular systems.
Collapse
Affiliation(s)
- Hui Ting Zhang
- European Molecular Biology Laboratory, 69117 Heidelberg, Germany;
| | - Takashi Hiiragi
- European Molecular Biology Laboratory, 69117 Heidelberg, Germany;
| |
Collapse
|
246
|
Chacón-Martínez CA, Koester J, Wickström SA. Signaling in the stem cell niche: regulating cell fate, function and plasticity. Development 2018; 145:145/15/dev165399. [PMID: 30068689 DOI: 10.1242/dev.165399] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Stem cells have the ability to self-renew and differentiate along multiple lineages, driving tissue homeostasis and regeneration. Paradigms of unidirectional, hierarchical differentiation trajectories observed in embryonic and hematopoietic stem cells have traditionally been applied to tissue-resident stem cells. However, accumulating evidence implicates stemness as a bidirectional, dynamic state that is largely governed by the niche, which facilitates plasticity and adaptability to changing conditions. In this Review, we discuss mechanisms of cell fate regulation through niche-derived cues, with a particular focus on epithelial stem cells of the mammalian skin, intestine and lung. We discuss a spectrum of niche-derived biochemical, mechanical and architectural inputs that define stem cell states during morphogenesis, homeostasis and regeneration, and highlight how these diverse inputs influence stem cell plasticity.
Collapse
Affiliation(s)
- Carlos Andrés Chacón-Martínez
- Paul Gerson Unna Group 'Skin Homeostasis and Ageing', Max Planck Institute for Biology of Ageing, D-50931 Cologne, Germany
| | - Janis Koester
- Paul Gerson Unna Group 'Skin Homeostasis and Ageing', Max Planck Institute for Biology of Ageing, D-50931 Cologne, Germany
| | - Sara A Wickström
- Paul Gerson Unna Group 'Skin Homeostasis and Ageing', Max Planck Institute for Biology of Ageing, D-50931 Cologne, Germany .,Helsinki Institute of Life Science, Biomedicum Helsinki, University of Helsinki, FI-00014 Helsinki, Finland.,Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
247
|
Bao M, Xie J, Huck WTS. Recent Advances in Engineering the Stem Cell Microniche in 3D. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800448. [PMID: 30128252 PMCID: PMC6096985 DOI: 10.1002/advs.201800448] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/01/2018] [Indexed: 05/18/2023]
Abstract
Conventional 2D cell culture techniques have provided fundamental insights into key biochemical and biophysical mechanisms responsible for various cellular behaviors, such as cell adhesion, spreading, division, proliferation, and differentiation. However, 2D culture in vitro does not fully capture the physical and chemical properties of the native microenvironment. There is a growing body of research that suggests that cells cultured on 2D substrates differ greatly from those grown in vivo. This article focuses on recent progress in using bioinspired 3D matrices that recapitulate as many aspects of the natural extracellular matrix as possible. A range of techniques for the engineering of 3D microenvironment with precisely controlled biophysical and chemical properties, and the impact of these environments on cellular behavior, is reviewed. Finally, an outlook on future challenges for engineering the 3D microenvironment and how such approaches would further our understanding of the influence of the microenvironment on cell function is provided.
Collapse
Affiliation(s)
- Min Bao
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525 AJNijmegenThe Netherlands
| | - Jing Xie
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525 AJNijmegenThe Netherlands
| | - Wilhelm T. S. Huck
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525 AJNijmegenThe Netherlands
| |
Collapse
|
248
|
Self-assembly of embryonic and two extra-embryonic stem cell types into gastrulating embryo-like structures. Nat Cell Biol 2018; 20:979-989. [PMID: 30038254 DOI: 10.1038/s41556-018-0147-7] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/20/2018] [Indexed: 02/08/2023]
Abstract
Embryonic stem cells can be incorporated into the developing embryo and its germ line, but, when cultured alone, their ability to generate embryonic structures is restricted. They can interact with trophoblast stem cells to generate structures that break symmetry and specify mesoderm, but their development is limited as the epithelial-mesenchymal transition of gastrulation cannot occur. Here, we describe a system that allows assembly of mouse embryonic, trophoblast and extra-embryonic endoderm stem cells into structures that acquire the embryo's architecture with all distinct embryonic and extra-embryonic compartments. Strikingly, such embryo-like structures develop to undertake the epithelial-mesenchymal transition, leading to mesoderm and then definitive endoderm specification. Spatial transcriptomic analyses demonstrate that these morphological transformations are underpinned by gene expression patterns characteristic of gastrulating embryos. This demonstrates the remarkable ability of three stem cell types to self-assemble in vitro into gastrulating embryo-like structures undertaking spatio-temporal events of the gastrulating mammalian embryo.
Collapse
|
249
|
Wörsdörfer P, Wagner N, Ergün S. The role of connexins during early embryonic development: pluripotent stem cells, gene editing, and artificial embryonic tissues as tools to close the knowledge gap. Histochem Cell Biol 2018; 150:327-339. [PMID: 30039329 DOI: 10.1007/s00418-018-1697-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2018] [Indexed: 12/14/2022]
Abstract
Since almost 4 decades, connexins have been discussed as important regulators of embryogenesis. Several different members of the gene family can be detected in the preimplantation embryo and during gastrulation. However, genetically engineered mice deficient for every connexin expressed during early development are available and even double-deficient mice were generated. Interestingly, all of these mice complete gastrulation without any abnormalities. This raises the question if the role of connexins has been overrated or if other gene family members compensate and mask their importance. To answer this question, embryos completely devoid of any gap junctional communication need to be investigated. This is challenging because a variety of connexin genes are co-expressed and some null mutations lead to a lethal phenotype. In addition, maternal connexin transcripts were described to persist until the blastocyst stage. In this review, we summarize the current knowledge about the role of connexins during preimplantation development and in embryonic stem cells. We propose that the use of pluripotent stem cells, trophoblast stem cells, as well as artificial embryo-like structures and organoid cultures in combination with multiplex CRISPR/Cas9-based genome editing provides a powerful platform to comprehensively readdress this issue and decipher the role of connexins during lineage decision, differentiation, and morphogenesis in a cell culture model for mouse and human development.
Collapse
Affiliation(s)
- Philipp Wörsdörfer
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstr.6, 97070, Würzburg, Germany.
| | - Nicole Wagner
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstr.6, 97070, Würzburg, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstr.6, 97070, Würzburg, Germany
| |
Collapse
|
250
|
Shahbazi MN, Zernicka-Goetz M. Deconstructing and reconstructing the mouse and human early embryo. Nat Cell Biol 2018; 20:878-887. [PMID: 30038253 DOI: 10.1038/s41556-018-0144-x] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 06/15/2018] [Indexed: 02/07/2023]
Abstract
The emergence of form and function during mammalian embryogenesis is a complex process that involves multiple regulatory levels. The foundations of the body plan are laid throughout the first days of post-implantation development as embryonic stem cells undergo symmetry breaking and initiate lineage specification, in a process that coincides with a global morphological reorganization of the embryo. Here, we review experimental models and how they have shaped our current understanding of the post-implantation mammalian embryo.
Collapse
Affiliation(s)
- Marta N Shahbazi
- Department of Physiology, Development and Neuroscience, Mammalian Embryo and Stem Cell Group, University of Cambridge, Cambridge, UK.
| | - Magdalena Zernicka-Goetz
- Department of Physiology, Development and Neuroscience, Mammalian Embryo and Stem Cell Group, University of Cambridge, Cambridge, UK.
| |
Collapse
|