1
|
Arias AM, Dias A, Wehmeyer AE, Arnold SJ, Fiúza UM. A modular organization of mammalian gastrulation and the Spemann-Mangold organizer. Cells Dev 2025:204031. [PMID: 40412479 DOI: 10.1016/j.cdev.2025.204031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 05/19/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Studies in amphibian embryos revealed the existence of groups of cells, organizers, that play a central role in laying down the body plan. One, the Spemann-Mangold Organizer (SMO) is associated with the induction of the nervous system and the development of the head, whereas a second one has been linked with the development of the trunk and the tail. Homologues of these organizers have been described in other vertebrates. In the mouse, the SMO organizer has been associated with a region of the mid- early gastrula and the tail-trunk Organizer with the node. Altogether these observations suggest a modular organization of the vertebrate body plan into three domains. One, most anterior, competent to form the brain, a middle one, associated with neural induction and the head, and one dedicated to axial extension. Work with gastruloids, pluripotent stem cell models of mammalian development, reveal that these modules are independent developmental units. Here we discuss the relationship of the gastruloids findings to the activity of organizers in embryos and the implications of this modular organization for the evolution of the vertebrate body plan.
Collapse
Affiliation(s)
- Alfonso Martinez Arias
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain; ICREA, Passeig de Lluís Companys, 23, L'Eixample, 08010 Barcelona, Spain
| | - André Dias
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Alexandra E Wehmeyer
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany
| | - Sebastian J Arnold
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany; Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Germany
| | - Ulla-Maj Fiúza
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
2
|
Marei HE. Stem Cell and Synthetic Embryo Models: Advances, Applications, and Ethical Considerations. Stem Cell Rev Rep 2025:10.1007/s12015-025-10890-z. [PMID: 40392412 DOI: 10.1007/s12015-025-10890-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2025] [Indexed: 05/22/2025]
Abstract
Independent traditional gametes and recent advances in stem cell biology have made it possible to create synthetic embryo models (SEMs), altering our capacity to study early human development, congenital diseases, and regenerative medicine. By recreating key developmental events in vitro, these models provide unmatched insights into embryogenesis and provide creative platforms for disease modeling, drug discovery, and individualized therapy. The quick development in SEM research raises serious ethical, legal, and regulatory questions that call for creating transparent control systems. The methods applied in SEM fabrication, their biomedical applications, and the moral issues connected with their use are investigated in this review. We also look at future directions, including enhancing ethical frameworks, adding artificial intelligence, increasing model fidelity, and encouraging public participation. Through multidisciplinary cooperation, SEMs might address these problems and transform developmental biology, advancing ethical scientific advancement.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35116, Egypt.
| |
Collapse
|
3
|
Villaronga-Luque A, Savill RG, López-Anguita N, Bolondi A, Garai S, Gassaloglu SI, Rouatbi R, Schmeisser K, Poddar A, Bauer L, Alves T, Traikov S, Rodenfels J, Chavakis T, Bulut-Karslioglu A, Veenvliet JV. Integrated molecular-phenotypic profiling reveals metabolic control of morphological variation in a stem-cell-based embryo model. Cell Stem Cell 2025; 32:759-777.e13. [PMID: 40245869 DOI: 10.1016/j.stem.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 11/27/2024] [Accepted: 03/21/2025] [Indexed: 04/19/2025]
Abstract
Considerable phenotypic variation under identical culture conditions limits the potential of stem-cell-based embryo models (SEMs) in basic and applied research. The biological processes causing this seemingly stochastic variation remain unclear. Here, we investigated the roots of phenotypic variation by parallel recording of transcriptomic states and morphological history in individual structures modeling embryonic trunk formation. Machine learning and integration of time-resolved single-cell RNA sequencing with imaging-based phenotypic profiling identified early features predictive of phenotypic end states. Leveraging this predictive power revealed that early imbalance of oxidative phosphorylation and glycolysis results in aberrant morphology and a neural lineage bias, which we confirmed by metabolic measurements. Accordingly, metabolic interventions improved phenotypic end states. Collectively, our work establishes divergent metabolic states as drivers of phenotypic variation and offers a broadly applicable framework to chart and predict phenotypic variation in organoids and SEMs. The strategy can be used to identify and control underlying biological processes, ultimately increasing reproducibility.
Collapse
Affiliation(s)
- Alba Villaronga-Luque
- Stembryogenesis Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Faculty of Biology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ryan G Savill
- Stembryogenesis Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Faculty of Biology, Technische Universität Dresden, 01307 Dresden, Germany
| | | | - Adriano Bolondi
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Sumit Garai
- Stembryogenesis Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01307 Dresden, Germany
| | - Seher Ipek Gassaloglu
- Stembryogenesis Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Roua Rouatbi
- MOSAIC Group, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Center for Systems Biology Dresden, 01307 Dresden, Germany; Faculty of Computer Science, Technische Universität Dresden, 01062 Dresden, Germany; Center for Scalable Data Analytics and Artificial Intelligence Dresden/Leipzig, 01062 Dresden, Germany
| | - Kathrin Schmeisser
- Energetics of Biological Systems Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Aayush Poddar
- Stembryogenesis Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Lisa Bauer
- Stembryogenesis Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Tiago Alves
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sofia Traikov
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jonathan Rodenfels
- Cluster of Excellence Physics of Life, Technische Universität Dresden, 01307 Dresden, Germany; Center for Systems Biology Dresden, 01307 Dresden, Germany; Energetics of Biological Systems Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | | | - Jesse V Veenvliet
- Stembryogenesis Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01307 Dresden, Germany; Center for Systems Biology Dresden, 01307 Dresden, Germany.
| |
Collapse
|
4
|
Rodríguez Colman MJ, Sonnen KF. Signaling switches: Metabolism regulates gastruloid self-organization. Cell Stem Cell 2025; 32:673-675. [PMID: 40315829 DOI: 10.1016/j.stem.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 04/07/2025] [Accepted: 04/07/2025] [Indexed: 05/04/2025]
Abstract
Metabolic regulation of embryonic development is increasingly recognized. Villaronga-Luque et al.1 and Stopornwongkul et al.2 show that metabolic activity influences gastruloid formation from mouse embryonic stem cells, revealing that the balance between glycolysis and oxidative phosphorylation regulates cell fate decisions during gastruloid self-organization.
Collapse
Affiliation(s)
- María J Rodríguez Colman
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands.
| | - Katharina F Sonnen
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
5
|
Haantjes RR, Strik J, de Visser J, Postma M, van Amerongen R, van Boxtel AL. Towards an integrated view and understanding of embryonic signalling during murine gastrulation. Cells Dev 2025:204028. [PMID: 40316255 DOI: 10.1016/j.cdev.2025.204028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
At the onset of mammalian gastrulation, secreted signalling molecules belonging to the Bmp, Wnt, Nodal and Fgf signalling pathways induce and pattern the primitive streak, marking the start for the cellular rearrangements that generate the body plan. Our current understanding of how signalling specifies and organises the germ layers in three dimensions, was mainly derived from genetic experimentation using mouse embryos performed over many decades. However, the exact spatiotemporal sequence of events is still poorly understood, both because of a lack of tractable models that allow for real time visualisation of signalling and differentiation and because of the molecular and cellular complexity of these early developmental events. In recent years, a new wave of in vitro embryo models has begun to shed light on the dynamics of signalling during primitive streak formation. Here we discuss the similarities and differences between a widely adopted mouse embryo model, termed gastruloids, and real embryos from a signalling perspective. We focus on the gene regulatory networks that underlie signalling pathway interactions and outline some of the challenges ahead. Finally, we provide a perspective on how embryo models may be used to advance our understanding of signalling dynamics through computational modelling.
Collapse
Affiliation(s)
- Rhanna R Haantjes
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands.
| | - Jeske Strik
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands; Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, 6525GA Nijmegen, the Netherlands.
| | - Joëlle de Visser
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands.
| | - Marten Postma
- Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands.
| | - Renée van Amerongen
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands.
| | - Antonius L van Boxtel
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands.
| |
Collapse
|
6
|
Amel A, Brown R, Rabeling A, Goolam M. Matrigel inhibits elongation and drives endoderm differentiation in aggregates of mouse embryonic stem cells. FEBS Open Bio 2025. [PMID: 40251891 DOI: 10.1002/2211-5463.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 04/05/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025] Open
Abstract
Modelling peri-implantation mammalian development using the self-organising properties of stem cells is a rapidly growing field that has advanced our understanding of cell fate decisions occurring in the early embryo. Matrigel, a basement membrane matrix, is a critical substrate used in various protocols for its efficacy in promoting stem cell growth and self-organisation. However, its role in driving stem cell lineage commitment, and whether this effect is driven by biochemical or physical cues, is not currently clear. Here, we grow embryoid bodies in suspension, Matrigel and agarose, an inert polysaccharide, to attempt to decouple the physical and biochemical roles of Matrigel and better understand how it drives stem cell differentiation. We use a combination of light microscopy, quantitative PCR and immunostaining to investigate gene and protein changes in our different culture conditions. We show that stem cell aggregates in Matrigel are hindered in their ability to elongate compared with those grown in agarose or in suspension, indicating that prohibitive role in self-organisation. Aggregates in Matrigel are also driven to differentiate into endoderm, with ectoderm differentiation inhibited. Furthermore, these effects are not due to the physical presence of Matrigel, as the same effects are not witnessed in aggregates grown in agarose. Our results thus indicate that Matrigel has a significant and complex effect on the differentiation and morphology of embryoid bodies.
Collapse
Affiliation(s)
- Atoosa Amel
- Department of Human Biology, University of Cape Town, South Africa
- UCT Neuroscience Institute, Cape Town, South Africa
| | - Rachel Brown
- Department of Human Biology, University of Cape Town, South Africa
- UCT Neuroscience Institute, Cape Town, South Africa
| | - Alexa Rabeling
- Department of Human Biology, University of Cape Town, South Africa
- UCT Neuroscience Institute, Cape Town, South Africa
| | - Mubeen Goolam
- Department of Human Biology, University of Cape Town, South Africa
- UCT Neuroscience Institute, Cape Town, South Africa
| |
Collapse
|
7
|
Neupane J, Lubatti G, Gross-Thebing T, Ruiz Tejada Segura ML, Butler R, Gross-Thebing S, Dietmann S, Scialdone A, Surani MA. The emergence of human primordial germ cell-like cells in stem cell-derived gastruloids. SCIENCE ADVANCES 2025; 11:eado1350. [PMID: 40138398 PMCID: PMC11939039 DOI: 10.1126/sciadv.ado1350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Most advances in early human postimplantation development depend on animal studies and stem cell-based embryo models. Here, we present self-organized three-dimensional human gastruloids (hGs) derived from embryonic stem cells. The transcriptome profile of day 3 hGs aligned with Carnegie stage 7 human gastrula, with cell types and differentiation trajectories consistent with human gastrulation. Notably, we observed the emergence of nascent primordial germ cell-like cells (PGCLCs), but without exogenous bone morphogenetic protein (BMP) signaling, which is essential for the PGCLC fate. A mutation in the ISL1 gene affects amnion-like cells and leads to a loss of PGCLCs; the addition of exogenous BMP2 rescues the PGCLC fate, indicating that the amnion may provide endogenous BMP signaling. Our model of early human embryogenesis will enable further exploration of the germ line and other early human lineages.
Collapse
Affiliation(s)
- Jitesh Neupane
- Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Physiology, Development and Neuroscience Department, University of Cambridge, Cambridge, UK
| | - Gabriele Lubatti
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, 81377 Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Functional Epigenetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Theresa Gross-Thebing
- Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Physiology, Development and Neuroscience Department, University of Cambridge, Cambridge, UK
| | - Mayra Luisa Ruiz Tejada Segura
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, 81377 Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Functional Epigenetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Richard Butler
- Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
| | | | - Sabine Dietmann
- Department of Development Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Antonio Scialdone
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, 81377 Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Functional Epigenetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - M. Azim Surani
- Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Physiology, Development and Neuroscience Department, University of Cambridge, Cambridge, UK
| |
Collapse
|
8
|
Blotenburg M, Suurenbroek L, Bax D, de Visser J, Bhardwaj V, Braccioli L, de Wit E, van Boxtel A, Marks H, Zeller P. Stem cell culture conditions affect in vitro differentiation potential and mouse gastruloid formation. PLoS One 2025; 20:e0317309. [PMID: 40138371 PMCID: PMC11940422 DOI: 10.1371/journal.pone.0317309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 12/24/2024] [Indexed: 03/29/2025] Open
Abstract
Aggregating low numbers of mouse embryonic stem cells (mESCs) and inducing Wnt signalling generates 'gastruloids', self-organising complex structures that display an anteroposterior organisation of cell types derived from all three germ layers. Current gastruloid protocols display considerable heterogeneity between experiments in terms of morphology, elongation efficiency, and cell type composition. We therefore investigated whether altering the mESC pluripotency state would provide more consistent results. By growing three mESC lines from two different genetic backgrounds in different intervals of ESLIF and 2i medium the pluripotency state of cells was modulated, and mESC culture as well as the resulting gastruloids were analysed. Microscopic analysis showed a pre-culture-specific effect on gastruloid formation, in terms of aspect ratio and reproducibility. RNA-seq analysis of the mESC start population confirmed that short-term pulses of 2i and ESLIF modulate the pluripotency state, and result in different cellular states. Since multiple epigenetic regulators were detected among the top differentially expressed genes, we further analysed genome-wide DNA methylation and H3K27me3 distributions. We observed epigenetic differences between conditions, most dominantly in the promoter regions of developmental regulators. Lastly, when we investigated the cell type composition of gastruloids grown from these different pre-cultures, we observed that mESCs subjected to 2i-ESLIF preceding aggregation generated gastruloids more consistently, including more complex mesodermal contributions as compared to the ESLIF-only control. These results indicate that optimisation of the mESCs pluripotency state allows the modulation of cell differentiation during gastruloid formation.
Collapse
Affiliation(s)
- Marloes Blotenburg
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Oncode Institute, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lianne Suurenbroek
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Oncode Institute, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Danique Bax
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Joëlle de Visser
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Vivek Bhardwaj
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Oncode Institute, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Luca Braccioli
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Elzo de Wit
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Antonius van Boxtel
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Hendrik Marks
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Peter Zeller
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Oncode Institute, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
9
|
Fu Y, Fan Q, Wu Y, Bao M. Unlocking the potential of stem-cell-derived 'synthetic' embryo models. Trends Biotechnol 2025:S0167-7799(25)00078-2. [PMID: 40090786 DOI: 10.1016/j.tibtech.2025.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 02/15/2025] [Accepted: 02/21/2025] [Indexed: 03/18/2025]
Abstract
Stem-cell-derived 'synthetic' embryo models represent a revolutionary avenue in developmental biology, offering unprecedented insights into embryogenesis and tissue formation. However, the majority of current research on embryo models resides predominantly in the engineering construction phase, with limited substantive applications. This review explores the utilization of these embryo models and their applications in deciphering fundamental developmental processes. We delve into the methodologies employed in generating these models, emphasizing their potential to advance our understanding of embryonic development and disease. By evaluating current advancements and challenges, this review provides a comprehensive overview of the opportunities and implications of employing stem-cell-derived embryo models.
Collapse
Affiliation(s)
- Yanqiong Fu
- OuJiang Laboratory, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Qin Fan
- OuJiang Laboratory, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yanru Wu
- OuJiang Laboratory, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Min Bao
- OuJiang Laboratory, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Department of Geriatric Medicine, First Affiliated Hospital of Wenzhou Medical Univesity, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
10
|
Braccioli L, van den Brand T, Alonso Saiz N, Fountas C, Celie PHN, Kazokaitė-Adomaitienė J, de Wit E. Identifying cross-lineage dependencies of cell-type-specific regulators in mouse gastruloids. Dev Cell 2025:S1534-5807(25)00118-2. [PMID: 40101716 DOI: 10.1016/j.devcel.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/09/2024] [Accepted: 02/21/2025] [Indexed: 03/20/2025]
Abstract
Correct gene expression levels are crucial for normal development. Advances in genomics enable the inference of gene regulatory programs active during development but cannot capture the complex multicellular interactions occurring during mammalian embryogenesis in utero. In vitro models of mammalian development, like gastruloids, can overcome this limitation. Using time-resolved single-cell chromatin accessibility analysis, we delineated the regulatory profile during mouse gastruloid development, identifying critical drivers of developmental transitions. Gastruloids develop from bipotent progenitor cells driven by the transcription factors (TFs) OCT4, SOX2, and TBXT, differentiating into the mesoderm (characterized by the mesogenin 1 [MSGN1]) and spinal cord (characterized by CDX2). ΔCDX gastruloids fail to form spinal cord, while Msgn1 ablation inhibits paraxial mesoderm and spinal cord development. Chimeric gastruloids with ΔMSGN1 and wild-type cells formed both tissues, indicating that inter-tissue communication is necessary for spinal cord formation. Our work has important implications for studying inter-tissue communication and gene regulatory programs in development.
Collapse
Affiliation(s)
- Luca Braccioli
- Division of Gene Regulation, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands.
| | - Teun van den Brand
- Division of Gene Regulation, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Noemi Alonso Saiz
- Division of Gene Regulation, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Charis Fountas
- Division of Gene Regulation, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Patrick H N Celie
- Protein Facility, Division of Biochemistry, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, 3521 AL Utrecht, the Netherlands
| | - Justina Kazokaitė-Adomaitienė
- Protein Facility, Division of Biochemistry, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, 3521 AL Utrecht, the Netherlands
| | - Elzo de Wit
- Division of Gene Regulation, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
11
|
Isomura A, Kageyama R. Progress in understanding the vertebrate segmentation clock. Nat Rev Genet 2025:10.1038/s41576-025-00813-6. [PMID: 40038453 DOI: 10.1038/s41576-025-00813-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2025] [Indexed: 03/06/2025]
Abstract
The segmentation clock is a molecular oscillator that regulates the periodic formation of somites from the presomitic mesoderm during vertebrate embryogenesis. Synchronous oscillatory expression of a Hairy homologue or Hairy-related basic helix-loop-helix (bHLH) transcriptional repressor in presomitic mesoderm cells regulates periodic expression of downstream factors that control somite segmentation with a periodicity that varies across species. Although many of the key components of the clock have been identified and characterized, less is known about how the clock is synchronized across cells and how species-specific periodicity is achieved. Advances in live imaging, stem cell and organoid technologies, and synthetic approaches have started to uncover the detailed mechanisms underlying these aspects of somitogenesis, providing insight into how morphogenesis is coordinated in space and time during embryonic development.
Collapse
Affiliation(s)
- Akihiro Isomura
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto, Japan.
- Japan Science and Technology Agency, PRESTO, Saitama, Japan.
- RIKEN Center for Brain Science, Wako, Japan.
| | - Ryoichiro Kageyama
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto, Japan.
- RIKEN Center for Brain Science, Wako, Japan.
| |
Collapse
|
12
|
Wu B, Neupane J, Zhou Y, Zhang J, Chen Y, Surani MA, Zhang Y, Bao S, Li X. Stem cell-based embryo models: a tool to study early human development. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2741-1. [PMID: 39969747 DOI: 10.1007/s11427-024-2741-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/23/2024] [Indexed: 02/20/2025]
Abstract
How a mammalian fertilized egg acquires totipotency and develops into a full-term offspring is a fundamental scientific question. Human embryonic development is difficult to study due to limited resources, technical challenges and ethics. Moreover, the precise regulatory mechanism underlying early human embryonic development remains unknown. In recent years, the emergence of stem cell-based embryo models (SCBEM) provides the opportunity to reconstitute pre- to post-implantation development in vitro. These models to some extent mimic the embryo morphologically and transcriptionally, and thus may be used to study key events in mammalian pre- and post-implantation development. Many groups have successfully generated SCBEM of the mouse and human. Here, we provide a comparative review of the mouse and human SCBEM, discuss the capability of these models to mimic natural embryos and give a perspective on their potential future applications.
Collapse
Affiliation(s)
- Baojiang Wu
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - Jitesh Neupane
- The Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Yang Zhou
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - Jingcheng Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Yanglin Chen
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - M Azim Surani
- The Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Yong Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China.
| | - Siqin Bao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
| | - Xihe Li
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
- Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic Animals, Hohhot, 011517, China.
| |
Collapse
|
13
|
Wang S, Shi G, Duan K, Yin Y, Li T. Extraembryonic mesoderm cells derived from human embryonic stem cells rely on Wnt pathway activation. Cell Prolif 2025; 58:e13761. [PMID: 39385268 PMCID: PMC11839190 DOI: 10.1111/cpr.13761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Extraembryonic mesoderm cells (EXMCs) are involved in the development of multiple embryonic lineages and umbilical cord formation, where they subsequently develop into mesenchymal stem cells (MSCs). Although EXMCs can be generated from human naïve embryonic stem cells (ESCs), it is unclear whether human primed ESCs (hpESCs) can differentiate into EXMCs that subsequently produce MSCs. The present report described a three-dimensional differentiation protocol to induce hpESCs into EXMCs by activating the Wnt pathway using CHIR99021. Single-cell transcriptome and immunostaining analyses revealed that the EXMC characteristics were similar to those of post-implantation embryonic EXMCs. Cell sorting was used to purify and expand the EXMCs. Importantly, these EXMCs secreted extracellular matrix proteins, including COL3A1 and differentiated into MSCs. Inconsistent with other MSC types, these MSCs exhibited a strong differentiation potential for chondrogenic and osteogenic cells and lacked adipocyte differentiation. Together, these findings provided a protocol to generate EXMCs and subsequent MSCs from hpESCs.
Collapse
Affiliation(s)
- Si‐Le Wang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnanChina
| | - Gao‐Hui Shi
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnanChina
| | - Kui Duan
- Department of Anatomy, College of Preclinical MedicineDali UniversityDaliYunnanChina
| | - Yu Yin
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnanChina
| | - Tianqing Li
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnanChina
| |
Collapse
|
14
|
Zhou Y, Tang C, Xiao X, Zhan X, Wang T, Xiao G, Xu L. Dimensionality reduction for visualizing spatially resolved profiling data using SpaSNE. Gigascience 2025; 14:giaf002. [PMID: 39960663 PMCID: PMC11831803 DOI: 10.1093/gigascience/giaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 11/05/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Spatially resolved profiling technologies to quantify transcriptomes, epigenomes, and proteomes have been emerging as groundbreaking methods for comprehensive molecular characterizations. Dimensionality reduction and visualization is an essential step to analyze and interpret spatially resolved profiling data. However, state-of-the-art dimensionality reduction methods for single-cell sequencing data, such as the t-distributed stochastic neighbor embedding (t-SNE) and uniform manifold approximation and projection (UMAP), were not tailored for spatially resolved profiling data. RESULTS Here we developed a spatially resolved t-SNE (SpaSNE) method to integrate both spatial and molecular information. We applied it to a variety of public spatially resolved profiling datasets that were generated from 3 experimental platforms and consisted of cells from different diseases, tissues, and cell types. To compare the performances of SpaSNE, t-SNE, and UMAP, we applied them to 4 spatially resolved profiling datasets obtained from 3 distinct experimental platforms (Visium, STARmap, and MERFISH) on both diseased and normal tissues. Comparisons between SpaSNE and these state-of-the-art approaches reveal that SpaSNE achieves more accurate and meaningful visualization that better elucidates the underlying spatial and molecular data structures. CONCLUSIONS This work demonstrates the broad application of SpaSNE for reliable and robust interpretation of cell types based on both molecular and spatial information, which can set the foundation for many subsequent analysis steps, such as differential gene expression and trajectory or pseudotime analysis on the spatially resolved profiling data.
Collapse
Affiliation(s)
- Yuansheng Zhou
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chen Tang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xue Xiao
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaowei Zhan
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tao Wang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Guanghua Xiao
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pediatrics, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
15
|
Magnitov MD, Maresca M, Alonso Saiz N, Teunissen H, Dong J, Sathyan KM, Braccioli L, Guertin MJ, de Wit E. ZNF143 is a transcriptional regulator of nuclear-encoded mitochondrial genes that acts independently of looping and CTCF. Mol Cell 2025; 85:24-41.e11. [PMID: 39708805 PMCID: PMC11687419 DOI: 10.1016/j.molcel.2024.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 09/23/2024] [Accepted: 11/22/2024] [Indexed: 12/23/2024]
Abstract
Gene expression is orchestrated by transcription factors, which function within the context of a three-dimensional genome. Zinc-finger protein 143 (ZNF143/ZFP143) is a transcription factor that has been implicated in both gene activation and chromatin looping. To study the direct consequences of ZNF143/ZFP143 loss, we generated a ZNF143/ZFP143 depletion system in mouse embryonic stem cells. Our results show that ZNF143/ZFP143 degradation has no effect on chromatin looping. Systematic analysis of ZNF143/ZFP143 occupancy data revealed that a commonly used antibody cross-reacts with CTCF, leading to its incorrect association with chromatin loops. Nevertheless, ZNF143/ZFP143 specifically activates nuclear-encoded mitochondrial genes, and its loss leads to severe mitochondrial dysfunction. Using an in vitro embryo model, we find that ZNF143/ZFP143 is an essential regulator of organismal development. Our results establish ZNF143/ZFP143 as a conserved transcriptional regulator of cell proliferation and differentiation by safeguarding mitochondrial activity.
Collapse
Affiliation(s)
- Mikhail D Magnitov
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Michela Maresca
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands; Department of Clinical Genetics, Erasmus University MC, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
| | - Noemí Alonso Saiz
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Hans Teunissen
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Jinhong Dong
- Center for Cell Analysis and Modeling, University of Connecticut, 400 Farmington Avenue, Farmington, CT, USA
| | - Kizhakke M Sathyan
- Center for Cell Analysis and Modeling, University of Connecticut, 400 Farmington Avenue, Farmington, CT, USA; Department of Genetics and Genome Sciences, University of Connecticut, 400 Farmington Avenue, Farmington, CT, USA
| | - Luca Braccioli
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Michael J Guertin
- Center for Cell Analysis and Modeling, University of Connecticut, 400 Farmington Avenue, Farmington, CT, USA; Department of Genetics and Genome Sciences, University of Connecticut, 400 Farmington Avenue, Farmington, CT, USA
| | - Elzo de Wit
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
16
|
Rohde LA, Bercowsky-Rama A, Valentin G, Naganathan SR, Desai RA, Strnad P, Soroldoni D, Oates AC. Cell-autonomous timing drives the vertebrate segmentation clock's wave pattern. eLife 2024; 13:RP93764. [PMID: 39671306 PMCID: PMC11643631 DOI: 10.7554/elife.93764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2024] Open
Abstract
Rhythmic and sequential segmentation of the growing vertebrate body relies on the segmentation clock, a multi-cellular oscillating genetic network. The clock is visible as tissue-level kinematic waves of gene expression that travel through the presomitic mesoderm (PSM) and arrest at the position of each forming segment. Here, we test how this hallmark wave pattern is driven by culturing single maturing PSM cells. We compare their cell-autonomous oscillatory and arrest dynamics to those we observe in the embryo at cellular resolution, finding similarity in the relative slowing of oscillations and arrest in concert with differentiation. This shows that cell-extrinsic signals are not required by the cells to instruct the developmental program underlying the wave pattern. We show that a cell-autonomous timing activity initiates during cell exit from the tailbud, then runs down in the anterior-ward cell flow in the PSM, thereby using elapsed time to provide positional information to the clock. Exogenous FGF lengthens the duration of the cell-intrinsic timer, indicating extrinsic factors in the embryo may regulate the segmentation clock via the timer. In sum, our work suggests that a noisy cell-autonomous, intrinsic timer drives the slowing and arrest of oscillations underlying the wave pattern, while extrinsic factors in the embryo tune this timer's duration and precision. This is a new insight into the balance of cell-intrinsic and -extrinsic mechanisms driving tissue patterning in development.
Collapse
Affiliation(s)
- Laurel A Rohde
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne EPFLLausanneSwitzerland
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Arianne Bercowsky-Rama
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne EPFLLausanneSwitzerland
| | - Guillaume Valentin
- Center of PhenoGenomics, Swiss Federal Institute of Technology in Lausanne EPFLLausanneSwitzerland
| | - Sundar Ram Naganathan
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne EPFLLausanneSwitzerland
- The Francis Crick InstituteLondonUnited Kingdom
| | - Ravi A Desai
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Petr Strnad
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne EPFLLausanneSwitzerland
| | - Daniele Soroldoni
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne EPFLLausanneSwitzerland
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Andrew C Oates
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne EPFLLausanneSwitzerland
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
- The Francis Crick InstituteLondonUnited Kingdom
| |
Collapse
|
17
|
Seubert AC, Krafft M, Bopp S, Helal M, Bhandare P, Wolf E, Alemany A, Riedel A, Kretzschmar K. Spatial transcriptomics reveals molecular cues underlying the site specificity of the adult mouse oral mucosa and its stem cell niches. Stem Cell Reports 2024; 19:1706-1719. [PMID: 39547226 PMCID: PMC11751799 DOI: 10.1016/j.stemcr.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
The oral cavity is a multifunctional organ composed of structurally heterogeneous mucosal tissues that remain poorly characterized. Oral mucosal tissues are highly stratified and segmented along an epithelial-lamina propria axis. Here, we performed spatial transcriptomics (tomo-seq) on the tongue, cheeks, and palate of the adult mouse to understand the cues that maintain the oral mucosal sites. We define molecular markers of unique and shared cellular niches and differentiation programs across oral sites. Using a comparative approach, we identify fibroblast growth factor (FGF) pathway components as potential stem cell niche factors for oral epithelial stem cells. Using organoid-forming efficiency assays, we validated three FGF ligands (FGF1, FGF7, and FGF10) as site-specific niche factors in the dorsal and ventral tongue. Our dataset of the spatially resolved genes across major oral sites represents a comprehensive resource for unraveling the molecular mechanisms underlying the adult homeostasis of the oral mucosa and its stem cell niches.
Collapse
Affiliation(s)
- Anna C Seubert
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, IZKF/MSNZ, University Hospital Würzburg, Würzburg, Germany; Graduate School of Life Sciences (GSLS), University of Würzburg, Würzburg, Germany
| | - Marion Krafft
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, IZKF/MSNZ, University Hospital Würzburg, Würzburg, Germany
| | - Sarah Bopp
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, IZKF/MSNZ, University Hospital Würzburg, Würzburg, Germany
| | - Moutaz Helal
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, IZKF/MSNZ, University Hospital Würzburg, Würzburg, Germany; Graduate School of Life Sciences (GSLS), University of Würzburg, Würzburg, Germany
| | | | - Elmar Wolf
- Institute of Biochemistry, Kiel University, Kiel, Germany
| | - Anna Alemany
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden, the Netherlands
| | - Angela Riedel
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, IZKF/MSNZ, University Hospital Würzburg, Würzburg, Germany; Graduate School of Life Sciences (GSLS), University of Würzburg, Würzburg, Germany
| | - Kai Kretzschmar
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, IZKF/MSNZ, University Hospital Würzburg, Würzburg, Germany; Graduate School of Life Sciences (GSLS), University of Würzburg, Würzburg, Germany.
| |
Collapse
|
18
|
Kokity L, Czimmerer Z, Benyhe-Kis B, Poscher A, Belai E, Steinbach G, Lipinszki Z, Pirity MK. Brachyury co-operates with polycomb protein RYBP to regulate gastrulation and axial elongation in vitro. Front Cell Dev Biol 2024; 12:1498346. [PMID: 39676794 PMCID: PMC11638158 DOI: 10.3389/fcell.2024.1498346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
Early embryonic development is a complex process where undifferentiated cells lose their pluripotency and start to gastrulate. During gastrulation, three germ layers form, giving rise to different cell lineages and organs. This process is regulated by transcription factors and epigenetic regulators, including non-canonical polycomb repressive complex 1s (ncPRC1s). Previously, we reported that ncPRC1-member RYBP (RING1 and YY1 binding protein) is crucial for embryonic implantation and cardiac lineage commitment in mice. However, the role of RYBP in gastrulation and mesoderm formation has not yet been defined. In this study, we used 2D and 3D in vitro model systems, to analyze the role of RYBP in mesoderm formation. First, we showed that cardiac and endothelial progenitors-both derived from mesoderm-are underrepresented in the Rybp -/- cardiac colonies. In the absence of RYBP, the formation of major germ layers was also disrupted, and the expression of mesoderm- (Brachyury, Eomes, and Gsc) and endoderm-specific (Sox17, Gata4) genes was significantly downregulated. Using 3D embryoid bodies as gastrulation models, we showed that RYBP can co-localize with mesoderm lineage marker protein BRACHYURY and endoderm marker protein GATA4 and both proteins. In mutants, both proteins were detected at low levels and showed altered distribution. Additionally, we compared our in vitro results to available in vivo single-cell transcriptomes and showed that Rybp and Brachyury co-expressed in the primitive streak and six mesodermal clusters. Since caudal mesoderm exhibited one of the strongest co-expressions, we tested axial elongation in wt and Rybp -/- gastruloids. In the absence of RYBP, gastruloids exhibited shortened tails and low BRACHYURY levels in the tailbud. Finally, we identified BRACHYURY as a novel binding partner of RYBP and presented evidence of possible cooperative function during mesoderm formation and axial elongation. Together, our results demonstrate the previously unknown role of RYBP in mesoderm formation. We believe our findings will contribute to better understanding of the highly conserved process of gastrulation.
Collapse
Affiliation(s)
- Lilla Kokity
- Biological Research Centre, Institute of Genetics, Hungarian Research Network, Szeged, Hungary
- Faculty of Science and Informatics, Doctoral School in Biology, University of Szeged, Szeged, Hungary
| | - Zsolt Czimmerer
- Biological Research Centre, Institute of Genetics, Hungarian Research Network, Szeged, Hungary
| | - Bernadett Benyhe-Kis
- Biological Research Centre, Institute of Genetics, Hungarian Research Network, Szeged, Hungary
- Faculty of Science and Informatics, Doctoral School in Biology, University of Szeged, Szeged, Hungary
| | - Anna Poscher
- Biological Research Centre, Institute of Genetics, Hungarian Research Network, Szeged, Hungary
- Faculty of Science and Informatics, Doctoral School in Biology, University of Szeged, Szeged, Hungary
| | - Emese Belai
- Biological Research Centre, Institute of Genetics, Hungarian Research Network, Szeged, Hungary
- Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Gábor Steinbach
- Cellular Imaging Laboratory, Core Facility, Biological Research Centre, Hungarian Research Network, Szeged, Hungary
| | - Zoltan Lipinszki
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Hungarian Research Network, Szeged, Hungary
- National Laboratory for Biotechnology, Institute of Genetics, Biological Research Centre, Hungarian Research Network, Szeged, Hungary
| | - Melinda Katalin Pirity
- Biological Research Centre, Institute of Genetics, Hungarian Research Network, Szeged, Hungary
| |
Collapse
|
19
|
Argiro L, Chevalier C, Choquet C, Nandkishore N, Ghata A, Baudot A, Zaffran S, Lescroart F. Gastruloids are competent to specify both cardiac and skeletal muscle lineages. Nat Commun 2024; 15:10172. [PMID: 39580459 PMCID: PMC11585638 DOI: 10.1038/s41467-024-54466-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/12/2024] [Indexed: 11/25/2024] Open
Abstract
Cardiopharyngeal mesoderm contributes to the formation of the heart and head muscles. However, the mechanisms governing cardiopharyngeal mesoderm specification remain unclear. Here, we reproduce cardiopharyngeal mesoderm specification towards cardiac and skeletal muscle lineages with gastruloids from mouse embryonic stem cells. By conducting a comprehensive temporal analysis of cardiopharyngeal mesoderm development and differentiation in gastruloids compared to mouse embryos, we present the evidence for skeletal myogenesis in gastruloids. We identify different subpopulations of cardiomyocytes and skeletal muscles, the latter of which most likely correspond to different states of myogenesis with "head-like" and "trunk-like" skeletal myoblasts. In this work, we unveil the potential of gastruloids to undergo specification into both cardiac and skeletal muscle lineages, allowing the investigation of the mechanisms of cardiopharyngeal mesoderm differentiation in development and how this could be affected in congenital diseases.
Collapse
Affiliation(s)
- Laurent Argiro
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Céline Chevalier
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Caroline Choquet
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Nitya Nandkishore
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
- Department of Biotechnology, SRM Institute of Science and Technology (SRMIST), Kattankulathur, Tamil Nadu, India
| | - Adeline Ghata
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Anaïs Baudot
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Stéphane Zaffran
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France.
| | - Fabienne Lescroart
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France.
| |
Collapse
|
20
|
Anlaş K, Gritti N, Nakaki F, Salamó Palau L, Tlili SL, Oriola D, Arató K, Le Lim J, Sharpe J, Trivedi V. Early autonomous patterning of the anteroposterior axis in gastruloids. Development 2024; 151:dev202171. [PMID: 39552366 DOI: 10.1242/dev.202171] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 06/17/2024] [Indexed: 11/19/2024]
Abstract
Minimal in vitro systems composed of embryonic stem cells (ESCs) have been shown to recapitulate the establishment of the anteroposterior (AP) axis. In contrast to the native embryo, ESC aggregates - such as gastruloids - can break symmetry, which is demarcated by polarization of the mesodermal marker T, autonomously without any localized external cues. However, associated earliest patterning events, such as the spatial restriction of cell fates and concomitant transcriptional changes, remain poorly understood. Here, we dissect the dynamics of AP axis establishment in mouse gastruloids, particularly before external Wnt stimulation. Through single-cell RNA sequencing, we identify key cell state transitions and the molecular signatures of T+ and T- populations underpinning AP polarization. We also show that this process is robust to modifications of aggregate size. Finally, transcriptomic comparison with the mouse embryo indicates that gastruloids develop similar mesendodermal cell types, despite initial differences in their primed pluripotent populations, which adopt a more mesenchymal state in lieu of an epiblast-like transcriptome. Hence, our findings suggest the possibility of alternate ESC states in vivo and in vitro that can converge onto similar cell fates.
Collapse
Affiliation(s)
| | | | | | | | - Sham Leilah Tlili
- Aix-Marseille Univ., CNRS, UMR 7288, IBDM, Turing Center for Living Systems, 13288 Marseille, France
| | | | | | | | - James Sharpe
- EMBL Barcelona, 08003 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| | - Vikas Trivedi
- EMBL Barcelona, 08003 Barcelona, Spain
- EMBL Heidelberg, Developmental Biology Unit, 69117 Heidelberg, Germany
| |
Collapse
|
21
|
McNamara HM, Solley SC, Adamson B, Chan MM, Toettcher JE. Recording morphogen signals reveals mechanisms underlying gastruloid symmetry breaking. Nat Cell Biol 2024; 26:1832-1844. [PMID: 39358450 PMCID: PMC11806519 DOI: 10.1038/s41556-024-01521-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 09/05/2024] [Indexed: 10/04/2024]
Abstract
Aggregates of stem cells can break symmetry and self-organize into embryo-like structures with complex morphologies and gene expression patterns. Mechanisms including reaction-diffusion Turing patterns and cell sorting have been proposed to explain symmetry breaking but distinguishing between these candidate mechanisms of self-organization requires identifying which early asymmetries evolve into subsequent tissue patterns and cell fates. Here we use synthetic 'signal-recording' gene circuits to trace the evolution of signalling patterns in gastruloids, three-dimensional stem cell aggregates that form an anterior-posterior axis and structures resembling the mammalian primitive streak and tailbud. We find that cell sorting rearranges patchy domains of Wnt activity into a single pole that defines the gastruloid anterior-posterior axis. We also trace the emergence of Wnt domains to earlier heterogeneity in Nodal activity even before Wnt activity is detectable. Our study defines a mechanism through which aggregates of stem cells can form a patterning axis even in the absence of external spatial cues.
Collapse
Affiliation(s)
- Harold M McNamara
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
| | - Sabrina C Solley
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Britt Adamson
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Michelle M Chan
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Jared E Toettcher
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
22
|
Cao D, Garai S, DiFrisco J, Veenvliet JV. The logic of monsters: development and morphological diversity in stem-cell-based embryo models. Interface Focus 2024; 14:20240023. [PMID: 39464644 PMCID: PMC11503023 DOI: 10.1098/rsfs.2024.0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 10/29/2024] Open
Abstract
Organoids and stem-cell-based embryo models (SEMs) are imperfect organ or embryo representations that explore a much larger space of possible forms, or morphospace, compared to their in vivo counterparts. Here, we discuss SEM biology in light of seminal work by Pere Alberch, a leading figure in early evo-devo, interpreting SEMs as developmental 'monstrosities' in the Alberchian sense. Alberch suggested that ordered patterns in aberrant development-i.e. 'the logic of monsters'-reveal developmental constraints on possible morphologies. In the same vein, we detail how SEMs have begun to shed light on structural features of normal development, such as developmental variability, the relative importance of internal versus external constraints, boundary conditions and design principles governing robustness and canalization. We argue that SEMs represent a powerful experimental tool to explore and expand developmental morphospace and propose that the 'monstrosity' of SEMs can be leveraged to uncover the 'hidden' rules and developmental constraints that robustly shape and pattern the embryo.
Collapse
Affiliation(s)
- Dominica Cao
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT06520, USA
| | - Sumit Garai
- Theoretical Biology Lab, The Francis Crick Institute, LondonNW1 1AT, UK
- Division of Biosciences, Medical Sciences Building, University College London, Gower Street, LondonWC1E 6BT, UK
| | - James DiFrisco
- Theoretical Biology Lab, The Francis Crick Institute, LondonNW1 1AT, UK
| | - Jesse V. Veenvliet
- Stembryogenesis Lab, Max Planck Institute of Molecular Cell Biology and Genetics, Dresden01307, Germany
- Center for Systems Biology Dresden, Dresden01307, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden01307, Germany
| |
Collapse
|
23
|
Schauer A, Veenvliet JV. Retinoid-enhanced human gastruloids. Nat Cell Biol 2024; 26:1634-1636. [PMID: 39394407 DOI: 10.1038/s41556-024-01517-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Affiliation(s)
- Alexandra Schauer
- Stembryogenesis Lab, Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Jesse V Veenvliet
- Stembryogenesis Lab, Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany.
- Center for Systems Biology Dresden, Dresden, Germany.
| |
Collapse
|
24
|
Hamazaki N, Yang W, Kubo CA, Qiu C, Martin BK, Garge RK, Regalado SG, Nichols EK, Pendyala S, Bradley N, Fowler DM, Lee C, Daza RM, Srivatsan S, Shendure J. Retinoic acid induces human gastruloids with posterior embryo-like structures. Nat Cell Biol 2024; 26:1790-1803. [PMID: 39164488 PMCID: PMC11469962 DOI: 10.1038/s41556-024-01487-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024]
Abstract
Gastruloids are a powerful in vitro model of early human development. However, although elongated and composed of all three germ layers, human gastruloids do not morphologically resemble post-implantation human embryos. Here we show that an early pulse of retinoic acid (RA), together with later Matrigel, robustly induces human gastruloids with posterior embryo-like morphological structures, including a neural tube flanked by segmented somites and diverse cell types, including neural crest, neural progenitors, renal progenitors and myocytes. Through in silico staging based on single-cell RNA sequencing, we find that human RA-gastruloids progress further than other human or mouse embryo models, aligning to E9.5 mouse and CS11 cynomolgus monkey embryos. We leverage chemical and genetic perturbations of RA-gastruloids to confirm that WNT and BMP signalling regulate somite formation and neural tube length in the human context, while transcription factors TBX6 and PAX3 underpin presomitic mesoderm and neural crest, respectively. Looking forward, RA-gastruloids are a robust, scalable model for decoding early human embryogenesis.
Collapse
Affiliation(s)
- Nobuhiko Hamazaki
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, USA.
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
- Seattle Hub for Synthetic Biology, Seattle, WA, USA.
| | - Wei Yang
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Connor A Kubo
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Chengxiang Qiu
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Beth K Martin
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Riddhiman K Garge
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Samuel G Regalado
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Eva K Nichols
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Sriram Pendyala
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Nicholas Bradley
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Douglas M Fowler
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Choli Lee
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Riza M Daza
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Sanjay Srivatsan
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
- Seattle Hub for Synthetic Biology, Seattle, WA, USA.
- Howard Hughes Medical Institute, Seattle, WA, USA.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
| |
Collapse
|
25
|
Li X, Zhu F, Min W. SpaDiT: diffusion transformer for spatial gene expression prediction using scRNA-seq. Brief Bioinform 2024; 25:bbae571. [PMID: 39508444 PMCID: PMC11541600 DOI: 10.1093/bib/bbae571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/24/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
The rapid development of spatially resolved transcriptomics (SRT) technologies has provided unprecedented opportunities for exploring the structure of specific organs or tissues. However, these techniques (such as image-based SRT) can achieve single-cell resolution, but can only capture the expression levels of tens to hundreds of genes. Such spatial transcriptomics (ST) data, carrying a large number of undetected genes, have limited its application value. To address the challenge, we develop SpaDiT, a deep learning framework for spatial reconstruction and gene expression prediction using scRNA-seq data. SpaDiT employs scRNA-seq data as an a priori condition and utilizes shared genes between ST and scRNA-seq data as latent representations to construct inputs, thereby facilitating the accurate prediction of gene expression in ST data. SpaDiT enhances the accuracy of spatial gene expression predictions over a variety of spatial transcriptomics datasets. We have demonstrated the effectiveness of SpaDiT by conducting extensive experiments on both seq-based and image-based ST data. We compared SpaDiT with eight highly effective baseline methods and found that our proposed method achieved an 8%-12% improvement in performance across multiple metrics. Source code and all datasets used in this paper are available at https://github.com/wenwenmin/SpaDiT and https://zenodo.org/records/12792074.
Collapse
Affiliation(s)
- Xiaoyu Li
- School of Information Science and Engineering, Yunnan University, 650500, Kunming, Yunnan, China
| | - Fangfang Zhu
- School of Health and Nursing, Yunnan Open University, 650599, Kunming, China
| | - Wenwen Min
- School of Information Science and Engineering, Yunnan University, 650500, Kunming, Yunnan, China
| |
Collapse
|
26
|
Farag N, Sacharen C, Avni L, Nachman I. Coordination between endoderm progression and mouse gastruloid elongation controls endodermal morphotype choice. Dev Cell 2024; 59:2364-2374.e4. [PMID: 38838673 DOI: 10.1016/j.devcel.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/11/2023] [Accepted: 05/15/2024] [Indexed: 06/07/2024]
Abstract
Embryonic development is highly robust. Morphogenetic variability between embryos (under ideal conditions) is largely quantitative. This robustness stands in contrast to in vitro embryo-like models, which, like most organoids, can display a high degree of tissue morphogenetic variability. The source of this difference is not fully understood. We use the mouse gastruloid model to study the morphogenetic progression of definitive endoderm (DE) and its divergence. We first catalog the different morphologies and characterize their statistics. We then learn predictive models for DE morphotype based on earlier expression and morphology measurements. Finally, we analyze these models to identify key drivers of morphotype variability and devise gastruloid-specific and global interventions that can lower this variability and steer morphotype choice. In the process, we identify two types of coordination lacking in the in vitro model but required for robust gut-tube formation. This approach can help improve the quality and usability of 3D embryo-like models.
Collapse
Affiliation(s)
- Naama Farag
- School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Chen Sacharen
- School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Lara Avni
- School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Iftach Nachman
- School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
27
|
Garge RK, Lynch V, Fields R, Casadei S, Best S, Stone J, Snyder M, McGann CD, Shendure J, Starita LM, Hamazaki N, Schweppe DK. The proteomic landscape and temporal dynamics of mammalian gastruloid development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.609098. [PMID: 39282277 PMCID: PMC11398484 DOI: 10.1101/2024.09.05.609098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Gastrulation is the highly coordinated process by which the early embryo breaks symmetry, establishes germ layers and a body plan, and sets the stage for organogenesis. As early mammalian development is challenging to study in vivo, stem cell-derived models have emerged as powerful surrogates, e.g. human and mouse gastruloids. However, although single cell RNA-seq (scRNA-seq) and high-resolution imaging have been extensively applied to characterize such in vitro embryo models, a paucity of measurements of protein dynamics and regulation leaves a major gap in our understanding. Here, we sought to address this by applying quantitative proteomics to human and mouse gastruloids at four key stages of their differentiation (naïve ESCs, primed ESCs, early gastruloids, late gastruloids). To the resulting data, we perform network analysis to map the dynamics of expression of macromolecular protein complexes and biochemical pathways, including identifying cooperative proteins that associate with them. With matched RNA-seq and phosphosite data from these same stages, we investigate pathway-, stage- and species-specific aspects of translational and post-translational regulation, e.g. finding peri-gastrulation stages of human and mice to be discordant with respect to the mitochondrial transcriptome vs. proteome, and nominating novel kinase-substrate relationships based on phosphosite dynamics. Finally, we leverage correlated dynamics to identify conserved protein networks centered around congenital disease genes. Altogether, our data (https://gastruloid.brotmanbaty.org/) and analyses showcase the potential of intersecting in vitro embryo models and proteomics to advance our understanding of early mammalian development in ways not possible through transcriptomics alone.
Collapse
Affiliation(s)
- Riddhiman K. Garge
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| | - Valerie Lynch
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Rose Fields
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Silvia Casadei
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| | - Sabrina Best
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| | - Jeremy Stone
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| | - Matthew Snyder
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| | - Chris D. McGann
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, Washington, USA
- Seattle Hub for Synthetic Biology, Seattle, Washington, USA
| | - Lea M. Starita
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| | - Nobuhiko Hamazaki
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
- Seattle Hub for Synthetic Biology, Seattle, Washington, USA
| | - Devin K. Schweppe
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
28
|
Miao Y, Pourquié O. Modeling human trunk development. Nat Biotechnol 2024; 42:1185-1186. [PMID: 37974011 DOI: 10.1038/s41587-023-02048-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Affiliation(s)
- Yuchuan Miao
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
29
|
Gribaudo S, Robert R, van Sambeek B, Mirdass C, Lyubimova A, Bouhali K, Ferent J, Morin X, van Oudenaarden A, Nedelec S. Self-organizing models of human trunk organogenesis recapitulate spinal cord and spine co-morphogenesis. Nat Biotechnol 2024; 42:1243-1253. [PMID: 37709912 DOI: 10.1038/s41587-023-01956-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 08/18/2023] [Indexed: 09/16/2023]
Abstract
Integrated in vitro models of human organogenesis are needed to elucidate the multi-systemic events underlying development and disease. Here we report the generation of human trunk-like structures that model the co-morphogenesis, patterning and differentiation of the human spine and spinal cord. We identified differentiation conditions for human pluripotent stem cells favoring the formation of an embryo-like extending antero-posterior (AP) axis. Single-cell and spatial transcriptomics show that somitic and spinal cord differentiation trajectories organize along this axis and can self-assemble into a neural tube surrounded by somites upon extracellular matrix addition. Morphogenesis is coupled with AP patterning mechanisms, which results, at later stages of organogenesis, in in vivo-like arrays of neural subtypes along a neural tube surrounded by spine and muscle progenitors contacted by neuronal projections. This integrated system of trunk development indicates that in vivo-like multi-tissue co-morphogenesis and topographic organization of terminal cell types can be achieved in human organoids, opening windows for the development of more complex models of organogenesis.
Collapse
Affiliation(s)
- Simona Gribaudo
- Institut du Fer à Moulin, Paris, France
- Inserm, UMR-S 1270, Paris, France
- Sorbonne Université, Science and Engineering Faculty, Paris, France
| | - Rémi Robert
- Institut du Fer à Moulin, Paris, France
- Inserm, UMR-S 1270, Paris, France
- Sorbonne Université, Science and Engineering Faculty, Paris, France
| | - Björn van Sambeek
- Oncode Institute, Utrecht, The Netherlands
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Camil Mirdass
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | - Anna Lyubimova
- Oncode Institute, Utrecht, The Netherlands
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kamal Bouhali
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Julien Ferent
- Institut du Fer à Moulin, Paris, France
- Inserm, UMR-S 1270, Paris, France
- Sorbonne Université, Science and Engineering Faculty, Paris, France
| | - Xavier Morin
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Alexander van Oudenaarden
- Oncode Institute, Utrecht, The Netherlands
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Stéphane Nedelec
- Institut du Fer à Moulin, Paris, France.
- Inserm, UMR-S 1270, Paris, France.
- Sorbonne Université, Science and Engineering Faculty, Paris, France.
| |
Collapse
|
30
|
Polak R, Zhang ET, Kuo CJ. Cancer organoids 2.0: modelling the complexity of the tumour immune microenvironment. Nat Rev Cancer 2024; 24:523-539. [PMID: 38977835 DOI: 10.1038/s41568-024-00706-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/09/2024] [Indexed: 07/10/2024]
Abstract
The development of neoplasia involves a complex and continuous interplay between malignantly transformed cells and the tumour microenvironment (TME). Cancer immunotherapies targeting the immune TME have been increasingly validated in clinical trials but response rates vary substantially between tumour histologies and are often transient, idiosyncratic and confounded by resistance. Faithful experimental models of the patient-specific tumour immune microenvironment, capable of recapitulating tumour biology and immunotherapy effects, would greatly improve patient selection, target identification and definition of resistance mechanisms for immuno-oncology therapeutics. In this Review, we discuss currently available and rapidly evolving 3D tumour organoid models that capture important immune features of the TME. We highlight diverse opportunities for organoid-based investigations of tumour immunity, drug development and precision medicine.
Collapse
Affiliation(s)
- Roel Polak
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Elisa T Zhang
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
31
|
Shoji JY, Davis RP, Mummery CL, Krauss S. Global Literature Analysis of Organoid and Organ-on-Chip Research. Adv Healthc Mater 2024; 13:e2301067. [PMID: 37479227 DOI: 10.1002/adhm.202301067] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/14/2023] [Indexed: 07/23/2023]
Abstract
Organoids and cells in organ-on-chip platforms replicate higher-level anatomical, physiological, or pathological states of tissues and organs. These technologies are widely regarded by academia, the pharmacological industry and regulators as key biomedical developments. To map advances in this emerging field, a literature analysis of 16,000 article metadata based on a quality-controlled text-mining algorithm is performed. The analysis covers titles, keywords, and abstracts of categorized academic publications in the literature and preprint databases published after 2010. The algorithm identifies and tracks 149 and 107 organs or organ substructures modeled as organoids and organ-on-chip, respectively, stem cell sources, as well as 130 diseases, and 16 groups of organisms other than human and mouse in which organoid/organ-on-chip technology is applied. The analysis illustrates changing diversity and focus in organoid/organ-on-chip research and captures its geographical distribution. The downloadable dataset provided is a robust framework for researchers to interrogate with their own questions.
Collapse
Affiliation(s)
- Jun-Ya Shoji
- Hybrid Technology Hub, Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0372, Norway
| | - Richard P Davis
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, 2300RC, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, 2300RC, the Netherlands
| | - Christine L Mummery
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, 2300RC, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, 2300RC, the Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, Enschede, 7522NB, the Netherlands
| | - Stefan Krauss
- Hybrid Technology Hub, Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0372, Norway
| |
Collapse
|
32
|
Chen W, Choi J, Li X, Nathans JF, Martin B, Yang W, Hamazaki N, Qiu C, Lalanne JB, Regalado S, Kim H, Agarwal V, Nichols E, Leith A, Lee C, Shendure J. Symbolic recording of signalling and cis-regulatory element activity to DNA. Nature 2024; 632:1073-1081. [PMID: 39020177 PMCID: PMC11357993 DOI: 10.1038/s41586-024-07706-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 06/12/2024] [Indexed: 07/19/2024]
Abstract
Measurements of gene expression or signal transduction activity are conventionally performed using methods that require either the destruction or live imaging of a biological sample within the timeframe of interest. Here we demonstrate an alternative paradigm in which such biological activities are stably recorded to the genome. Enhancer-driven genomic recording of transcriptional activity in multiplex (ENGRAM) is based on the signal-dependent production of prime editing guide RNAs that mediate the insertion of signal-specific barcodes (symbols) into a genomically encoded recording unit. We show how this strategy can be used for multiplex recording of the cell-type-specific activities of dozens to hundreds of cis-regulatory elements with high fidelity, sensitivity and reproducibility. Leveraging signal transduction pathway-responsive cis-regulatory elements, we also demonstrate time- and concentration-dependent genomic recording of WNT, NF-κB and Tet-On activities. By coupling ENGRAM to sequential genome editing via DNA Typewriter1, we stably record information about the temporal dynamics of two orthogonal signalling pathways to genomic DNA. Finally we apply ENGRAM to integratively record the transient activity of nearly 100 transcription factor consensus motifs across daily windows spanning the differentiation of mouse embryonic stem cells into gastruloids, an in vitro model of early mammalian development. Although these are proof-of-concept experiments and much work remains to fully realize the possibilities, the symbolic recording of biological signals or states within cells, to the genome and over time, has broad potential to complement contemporary paradigms for how we make measurements in biological systems.
Collapse
Affiliation(s)
- Wei Chen
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA.
| | - Junhong Choi
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Xiaoyi Li
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Jenny F Nathans
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Beth Martin
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Wei Yang
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Nobuhiko Hamazaki
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, USA
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA
| | - Chengxiang Qiu
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | | | - Samuel Regalado
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Haedong Kim
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Vikram Agarwal
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Eva Nichols
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Anh Leith
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Choli Lee
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Howard Hughes Medical Institute, Seattle, WA, USA.
- Seattle Hub for Synthetic Biology, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
| |
Collapse
|
33
|
Rebuzzini P, Rustichelli S, Fassina L, Canobbio I, Zuccotti M, Garagna S. BPA Exposure Affects Mouse Gastruloids Axial Elongation by Perturbing the Wnt/β-Catenin Pathway. Int J Mol Sci 2024; 25:7924. [PMID: 39063166 PMCID: PMC11276681 DOI: 10.3390/ijms25147924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Mammalian embryos are very vulnerable to environmental toxicants (ETs) exposure. Bisphenol A (BPA), one of the most diffused ETs, exerts endocrine-disrupting effects through estro-gen-mimicking and hormone-like properties, with detrimental health effects, including on reproduction. However, its impact during the peri-implantation stages is still unclear. This study, using gastruloids as a 3D stem cell-based in vitro model of embryonic development, showed that BPA exposure arrests their axial elongation when present during the Wnt/β-catenin pathway activation period by β-catenin protein reduction. Gastruloid reshaping might have been impeded by the downregulation of Snail, Slug and Twist, known to suppress E-cadherin expression and to activate the N-cadherin gene, and by the low expression of the N-cadherin protein. Also, the lack of gastruloids elongation might be related to altered exit of BPA-exposed cells from the pluripotency condition and their following differentiation. In conclusion, here we show that the inhibition of gastruloids' axial elongation by BPA might be the result of the concomitant Wnt/β-catenin perturbation, reduced N-cadherin expression and Oct4, T/Bra and Cdx2 altered patter expression, which all together concur in the impaired development of mouse gastruloids.
Collapse
Affiliation(s)
- Paola Rebuzzini
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy; (M.Z.); (S.G.)
| | - Serena Rustichelli
- Laboratory of Biochemistry, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Via Bassi 21, 27100 Pavia, Italy; (S.R.); (I.C.)
- University School for Advanced Studies Pavia (IUSS), 27100 Pavia, Italy
| | - Lorenzo Fassina
- Department of Electrical, Computer and Biomedical Engineering (DIII), University of Pavia, Via Ferrata 5, 27100 Pavia, Italy;
- Centre for Health Technologies (CHT), University of Pavia, Via Ferrata 5, 27100 Pavia, Italy
| | - Ilaria Canobbio
- Laboratory of Biochemistry, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Via Bassi 21, 27100 Pavia, Italy; (S.R.); (I.C.)
| | - Maurizio Zuccotti
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy; (M.Z.); (S.G.)
- Centre for Health Technologies (CHT), University of Pavia, Via Ferrata 5, 27100 Pavia, Italy
| | - Silvia Garagna
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy; (M.Z.); (S.G.)
- Centre for Health Technologies (CHT), University of Pavia, Via Ferrata 5, 27100 Pavia, Italy
| |
Collapse
|
34
|
Stelloo S, Alejo-Vinogradova MT, van Gelder CAGH, Zijlmans DW, van Oostrom MJ, Valverde JM, Lamers LA, Rus T, Sobrevals Alcaraz P, Schäfers T, Furlan C, Jansen PWTC, Baltissen MPA, Sonnen KF, Burgering B, Altelaar MAFM, Vos HR, Vermeulen M. Deciphering lineage specification during early embryogenesis in mouse gastruloids using multilayered proteomics. Cell Stem Cell 2024; 31:1072-1090.e8. [PMID: 38754429 DOI: 10.1016/j.stem.2024.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 01/10/2024] [Accepted: 04/19/2024] [Indexed: 05/18/2024]
Abstract
Gastrulation is a critical stage in embryonic development during which the germ layers are established. Advances in sequencing technologies led to the identification of gene regulatory programs that control the emergence of the germ layers and their derivatives. However, proteome-based studies of early mammalian development are scarce. To overcome this, we utilized gastruloids and a multilayered mass spectrometry-based proteomics approach to investigate the global dynamics of (phospho) protein expression during gastruloid differentiation. Our findings revealed many proteins with temporal expression and unique expression profiles for each germ layer, which we also validated using single-cell proteomics technology. Additionally, we profiled enhancer interaction landscapes using P300 proximity labeling, which revealed numerous gastruloid-specific transcription factors and chromatin remodelers. Subsequent degron-based perturbations combined with single-cell RNA sequencing (scRNA-seq) identified a critical role for ZEB2 in mouse and human somitogenesis. Overall, this study provides a rich resource for developmental and synthetic biology communities endeavoring to understand mammalian embryogenesis.
Collapse
Affiliation(s)
- Suzan Stelloo
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525 GA Nijmegen, the Netherlands.
| | - Maria Teresa Alejo-Vinogradova
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525 GA Nijmegen, the Netherlands
| | - Charlotte A G H van Gelder
- Molecular Cancer Research, Center for Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Dick W Zijlmans
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525 GA Nijmegen, the Netherlands
| | - Marek J van Oostrom
- Hubrecht Institute, KNAW (Royal Netherlands Academy of Arts and Sciences), University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands
| | - Juan Manuel Valverde
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CA Utrecht, the Netherlands; Netherlands Proteomics Center, 3584 CH Utrecht, the Netherlands
| | - Lieke A Lamers
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525 GA Nijmegen, the Netherlands
| | - Teja Rus
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525 GA Nijmegen, the Netherlands
| | - Paula Sobrevals Alcaraz
- Molecular Cancer Research, Center for Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Tilman Schäfers
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, 6525 GA Nijmegen, the Netherlands
| | - Cristina Furlan
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, 6708 WE Wageningen, the Netherlands
| | - Pascal W T C Jansen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525 GA Nijmegen, the Netherlands
| | - Marijke P A Baltissen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525 GA Nijmegen, the Netherlands
| | - Katharina F Sonnen
- Hubrecht Institute, KNAW (Royal Netherlands Academy of Arts and Sciences), University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands
| | - Boudewijn Burgering
- Molecular Cancer Research, Center for Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Maarten A F M Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CA Utrecht, the Netherlands; Netherlands Proteomics Center, 3584 CH Utrecht, the Netherlands
| | - Harmjan R Vos
- Molecular Cancer Research, Center for Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525 GA Nijmegen, the Netherlands; Division of Molecular Genetics, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
35
|
Miao Y, Pourquié O. Cellular and molecular control of vertebrate somitogenesis. Nat Rev Mol Cell Biol 2024; 25:517-533. [PMID: 38418851 PMCID: PMC11694818 DOI: 10.1038/s41580-024-00709-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2024] [Indexed: 03/02/2024]
Abstract
Segmentation is a fundamental feature of the vertebrate body plan. This metameric organization is first implemented by somitogenesis in the early embryo, when paired epithelial blocks called somites are rhythmically formed to flank the neural tube. Recent advances in in vitro models have offered new opportunities to elucidate the mechanisms that underlie somitogenesis. Notably, models derived from human pluripotent stem cells introduced an efficient proxy for studying this process during human development. In this Review, we summarize the current understanding of somitogenesis gained from both in vivo studies and in vitro studies. We deconstruct the spatiotemporal dynamics of somitogenesis into four distinct modules: dynamic events in the presomitic mesoderm, segmental determination, somite anteroposterior polarity patterning, and epithelial morphogenesis. We first focus on the segmentation clock, as well as signalling and metabolic gradients along the tissue, before discussing the clock and wavefront and other models that account for segmental determination. We then detail the molecular and cellular mechanisms of anteroposterior polarity patterning and somite epithelialization.
Collapse
Affiliation(s)
- Yuchuan Miao
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
36
|
Wu J, Fu J. Toward developing human organs via embryo models and chimeras. Cell 2024; 187:3194-3219. [PMID: 38906095 PMCID: PMC11239105 DOI: 10.1016/j.cell.2024.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 02/02/2024] [Accepted: 05/14/2024] [Indexed: 06/23/2024]
Abstract
Developing functional organs from stem cells remains a challenging goal in regenerative medicine. Existing methodologies, such as tissue engineering, bioprinting, and organoids, only offer partial solutions. This perspective focuses on two promising approaches emerging for engineering human organs from stem cells: stem cell-based embryo models and interspecies organogenesis. Both approaches exploit the premise of guiding stem cells to mimic natural development. We begin by summarizing what is known about early human development as a blueprint for recapitulating organogenesis in both embryo models and interspecies chimeras. The latest advances in both fields are discussed before highlighting the technological and knowledge gaps to be addressed before the goal of developing human organs could be achieved using the two approaches. We conclude by discussing challenges facing embryo modeling and interspecies organogenesis and outlining future prospects for advancing both fields toward the generation of human tissues and organs for basic research and translational applications.
Collapse
Affiliation(s)
- Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
37
|
Dingare C, Cao D, Yang JJ, Sozen B, Steventon B. Mannose controls mesoderm specification and symmetry breaking in mouse gastruloids. Dev Cell 2024; 59:1523-1537.e6. [PMID: 38636516 DOI: 10.1016/j.devcel.2024.03.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 01/29/2024] [Accepted: 03/21/2024] [Indexed: 04/20/2024]
Abstract
Patterning and growth are fundamental features of embryonic development that must be tightly coordinated. To understand how metabolism impacts early mesoderm development, we used mouse embryonic stem-cell-derived gastruloids, that co-expressed glucose transporters with the mesodermal marker T/Bra. We found that the glucose mimic, 2-deoxy-D-glucose (2-DG), blocked T/Bra expression and abolished axial elongation in gastruloids. However, glucose removal did not phenocopy 2-DG treatment despite a decline in glycolytic intermediates. As 2-DG can also act as a competitive inhibitor of mannose in protein glycosylation, we added mannose together with 2-DG and found that it could rescue the mesoderm specification both in vivo and in vitro. We further showed that blocking production and intracellular recycling of mannose abrogated mesoderm specification. Proteomics analysis demonstrated that mannose reversed glycosylation of the Wnt pathway regulator, secreted frizzled receptor Frzb. Our study showed how mannose controls mesoderm specification in mouse gastruloids.
Collapse
Affiliation(s)
- Chaitanya Dingare
- Deptartment of Genetics, University of Cambridge, Downing Site, Cambridge CB2 3EH, UK.
| | - Dominica Cao
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Jenny Jingni Yang
- Deptartment of Genetics, University of Cambridge, Downing Site, Cambridge CB2 3EH, UK
| | - Berna Sozen
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA; Yale Stem Cell Centre, Yale University, New Haven, CT, USA; Department of Obstetrics, Gynaecology and Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Benjamin Steventon
- Deptartment of Genetics, University of Cambridge, Downing Site, Cambridge CB2 3EH, UK.
| |
Collapse
|
38
|
Merle M, Friedman L, Chureau C, Shoushtarizadeh A, Gregor T. Precise and scalable self-organization in mammalian pseudo-embryos. Nat Struct Mol Biol 2024; 31:896-902. [PMID: 38491138 DOI: 10.1038/s41594-024-01251-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 02/08/2024] [Indexed: 03/18/2024]
Abstract
Gene expression is inherently noisy, posing a challenge to understanding how precise and reproducible patterns of gene expression emerge in mammals. Here we investigate this phenomenon using gastruloids, a three-dimensional in vitro model for early mammalian development. Our study reveals intrinsic reproducibility in the self-organization of gastruloids, encompassing growth dynamics and gene expression patterns. We observe a remarkable degree of control over gene expression along the main body axis, with pattern boundaries positioned with single-cell precision. Furthermore, as gastruloids grow, both their physical proportions and gene expression patterns scale proportionally with system size. Notably, these properties emerge spontaneously in self-organizing cell aggregates, distinct from many in vivo systems constrained by fixed boundary conditions. Our findings shed light on the intricacies of developmental precision, reproducibility and size scaling within a mammalian system, suggesting that these phenomena might constitute fundamental features of multicellularity.
Collapse
Affiliation(s)
- Mélody Merle
- Department of Developmental and Stem Cell Biology, CNRS UMR3738 Paris Cité, Institut Pasteur, Paris, France
| | - Leah Friedman
- Department of Developmental and Stem Cell Biology, CNRS UMR3738 Paris Cité, Institut Pasteur, Paris, France
| | - Corinne Chureau
- Department of Developmental and Stem Cell Biology, CNRS UMR3738 Paris Cité, Institut Pasteur, Paris, France
| | - Armin Shoushtarizadeh
- Department of Developmental and Stem Cell Biology, CNRS UMR3738 Paris Cité, Institut Pasteur, Paris, France
| | - Thomas Gregor
- Department of Developmental and Stem Cell Biology, CNRS UMR3738 Paris Cité, Institut Pasteur, Paris, France.
- Joseph Henry Laboratories of Physics & Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
39
|
Fujiwara N, Kimura G, Nakagawa H. Emerging Roles of Spatial Transcriptomics in Liver Research. Semin Liver Dis 2024; 44:115-132. [PMID: 38574750 DOI: 10.1055/a-2299-7880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Spatial transcriptomics, leveraging sequencing- and imaging-based techniques, has emerged as a groundbreaking technology for mapping gene expression within the complex architectures of tissues. This approach provides an in-depth understanding of cellular and molecular dynamics across various states of healthy and diseased livers. Through the integration of sophisticated bioinformatics strategies, it enables detailed exploration of cellular heterogeneity, transitions in cell states, and intricate cell-cell interactions with remarkable precision. In liver research, spatial transcriptomics has been particularly revelatory, identifying distinct zonated functions of hepatocytes that are crucial for understanding the metabolic and detoxification processes of the liver. Moreover, this technology has unveiled new insights into the pathogenesis of liver diseases, such as the role of lipid-associated macrophages in steatosis and endothelial cell signals in liver regeneration and repair. In the domain of liver cancer, spatial transcriptomics has proven instrumental in delineating intratumor heterogeneity, identifying supportive microenvironmental niches and revealing the complex interplay between tumor cells and the immune system as well as susceptibility to immune checkpoint inhibitors. In conclusion, spatial transcriptomics represents a significant advance in hepatology, promising to enhance our understanding and treatment of liver diseases.
Collapse
Affiliation(s)
- Naoto Fujiwara
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Mie University, Mie, Japan
| | - Genki Kimura
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Mie University, Mie, Japan
| | - Hayato Nakagawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Mie University, Mie, Japan
| |
Collapse
|
40
|
Zhou G, Pang S, Li Y, Gao J. Progress in the generation of spinal cord organoids over the past decade and future perspectives. Neural Regen Res 2024; 19:1013-1019. [PMID: 37862203 PMCID: PMC10749595 DOI: 10.4103/1673-5374.385280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/25/2023] [Accepted: 08/01/2023] [Indexed: 10/22/2023] Open
Abstract
Spinal cord organoids are three-dimensional tissues derived from stem cells that recapitulate the primary morphological and functional characteristics of the spinal cord in vivo. As emerging bioengineering methods have led to the optimization of cell culture protocols, spinal cord organoids technology has made remarkable advancements in the past decade. Our literature search found that current spinal cord organoids do not only dynamically simulate neural tube formation but also exhibit diverse cytoarchitecture along the dorsal-ventral and rostral-caudal axes. Moreover, fused organoids that integrate motor neurons and other regionally specific organoids exhibit intricate neural circuits that allows for functional assessment. These qualities make spinal cord organoids valuable tools for disease modeling, drug screening, and tissue regeneration. By utilizing this emergent technology, researchers have made significant progress in investigating the pathogenesis and potential therapeutic targets of spinal cord diseases. However, at present, spinal cord organoid technology remains in its infancy and has not been widely applied in translational medicine. Establishment of the next generation of spinal cord organoids will depend on good manufacturing practice standards and needs to focus on diverse cell phenotypes and electrophysiological functionality evaluation.
Collapse
Affiliation(s)
- Gang Zhou
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Siyuan Pang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yongning Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of International Medical Service, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jun Gao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
41
|
McMillen P, Levin M. Collective intelligence: A unifying concept for integrating biology across scales and substrates. Commun Biol 2024; 7:378. [PMID: 38548821 PMCID: PMC10978875 DOI: 10.1038/s42003-024-06037-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 03/11/2024] [Indexed: 04/01/2024] Open
Abstract
A defining feature of biology is the use of a multiscale architecture, ranging from molecular networks to cells, tissues, organs, whole bodies, and swarms. Crucially however, biology is not only nested structurally, but also functionally: each level is able to solve problems in distinct problem spaces, such as physiological, morphological, and behavioral state space. Percolating adaptive functionality from one level of competent subunits to a higher functional level of organization requires collective dynamics: multiple components must work together to achieve specific outcomes. Here we overview a number of biological examples at different scales which highlight the ability of cellular material to make decisions that implement cooperation toward specific homeodynamic endpoints, and implement collective intelligence by solving problems at the cell, tissue, and whole-organism levels. We explore the hypothesis that collective intelligence is not only the province of groups of animals, and that an important symmetry exists between the behavioral science of swarms and the competencies of cells and other biological systems at different scales. We then briefly outline the implications of this approach, and the possible impact of tools from the field of diverse intelligence for regenerative medicine and synthetic bioengineering.
Collapse
Affiliation(s)
- Patrick McMillen
- Department of Biology, Tufts University, Medford, MA, 02155, USA
- Allen Discovery Center at Tufts University, Medford, MA, 02155, USA
| | - Michael Levin
- Department of Biology, Tufts University, Medford, MA, 02155, USA.
- Allen Discovery Center at Tufts University, Medford, MA, 02155, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
| |
Collapse
|
42
|
Martins-Costa C, Wilson V, Binagui-Casas A. Neuromesodermal specification during head-to-tail body axis formation. Curr Top Dev Biol 2024; 159:232-271. [PMID: 38729677 DOI: 10.1016/bs.ctdb.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
The anterior-to-posterior (head-to-tail) body axis is extraordinarily diverse among vertebrates but conserved within species. Body axis development requires a population of axial progenitors that resides at the posterior of the embryo to sustain elongation and is then eliminated once axis extension is complete. These progenitors occupy distinct domains in the posterior (tail-end) of the embryo and contribute to various lineages along the body axis. The subset of axial progenitors with neuromesodermal competency will generate both the neural tube (the precursor of the spinal cord), and the trunk and tail somites (producing the musculoskeleton) during embryo development. These axial progenitors are called Neuromesodermal Competent cells (NMCs) and Neuromesodermal Progenitors (NMPs). NMCs/NMPs have recently attracted interest beyond the field of developmental biology due to their clinical potential. In the mouse, the maintenance of neuromesodermal competency relies on a fine balance between a trio of known signals: Wnt/β-catenin, FGF signalling activity and suppression of retinoic acid signalling. These signals regulate the relative expression levels of the mesodermal transcription factor Brachyury and the neural transcription factor Sox2, permitting the maintenance of progenitor identity when co-expressed, and either mesoderm or neural lineage commitment when the balance is tilted towards either Brachyury or Sox2, respectively. Despite important advances in understanding key genes and cellular behaviours involved in these fate decisions, how the balance between mesodermal and neural fates is achieved remains largely unknown. In this chapter, we provide an overview of signalling and gene regulatory networks in NMCs/NMPs. We discuss mutant phenotypes associated with axial defects, hinting at the potential significant role of lesser studied proteins in the maintenance and differentiation of the progenitors that fuel axial elongation.
Collapse
Affiliation(s)
- C Martins-Costa
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
| | - V Wilson
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom.
| | - A Binagui-Casas
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
43
|
Rodriguez-Polo I, Moris N. Using Embryo Models to Understand the Development and Progression of Embryonic Lineages: A Focus on Primordial Germ Cell Development. Cells Tissues Organs 2024; 213:503-522. [PMID: 38479364 PMCID: PMC7616515 DOI: 10.1159/000538275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/05/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Recapitulating mammalian cell type differentiation in vitro promises to improve our understanding of how these processes happen in vivo, while bringing additional prospects for biomedical applications. The establishment of stem cell-derived embryo models and embryonic organoids, which have experienced explosive growth over the last few years, opens new avenues for research due to their scale, reproducibility, and accessibility. Embryo models mimic various developmental stages, exhibit different degrees of complexity, and can be established across species. Since embryo models exhibit multiple lineages organized spatially and temporally, they are likely to provide cellular niches that, to some degree, recapitulate the embryonic setting and enable "co-development" between cell types and neighbouring populations. One example where this is already apparent is in the case of primordial germ cell-like cells (PGCLCs). SUMMARY While directed differentiation protocols enable the efficient generation of high PGCLC numbers, embryo models provide an attractive alternative as they enable the study of interactions of PGCLCs with neighbouring cells, alongside the regulatory molecular and biophysical mechanisms of PGC competency. Additionally, some embryo models can recapitulate post-specification stages of PGC development (including migration or gametogenesis), mimicking the inductive signals pushing PGCLCs to mature and differentiate and enabling the study of PGCLC development across stages. Therefore, in vitro models may allow us to address questions of cell type differentiation, and PGC development specifically, that have hitherto been out of reach with existing systems. KEY MESSAGE This review evaluates the current advances in stem cell-based embryo models, with a focus on their potential to model cell type-specific differentiation in general and in particular to address open questions in PGC development and gametogenesis.
Collapse
Affiliation(s)
| | - Naomi Moris
- The Francis Crick Institute, 1 Midland Road, Somers Town, London, NW1 1AT, UK
| |
Collapse
|
44
|
Hamazaki N, Yang W, Kubo C, Qiu C, Martin BK, Garge RK, Regalado SG, Nichols E, Lee C, Daza RM, Srivatsan S, Shendure J. Induction and in silico staging of human gastruloids with neural tube, segmented somites & advanced cell types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.10.579769. [PMID: 38405970 PMCID: PMC10888963 DOI: 10.1101/2024.02.10.579769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Embryonic organoids are emerging as powerful models for studying early mammalian development. For example, stem cell-derived 'gastruloids' form elongating structures containing all three germ layers1-4. However, although elongated, human gastruloids do not morphologically resemble post-implantation embryos. Here we show that a specific, discontinuous regimen of retinoic acid (RA) robustly induces human gastruloids with embryo-like morphological structures, including a neural tube and segmented somites. Single cell RNA-seq (sc-RNA-seq) further reveals that these human 'RA-gastruloids' contain more advanced cell types than conventional gastruloids, including neural crest cells, renal progenitor cells, skeletal muscle cells, and, rarely, neural progenitor cells. We apply a new approach to computationally stage human RA-gastruloids relative to somite-resolved mouse embryos, early human embryos and other gastruloid models, and find that the developmental stage of human RA-gastruloids is comparable to that of E9.5 mouse embryos, although some cell types show greater or lesser progression. We chemically perturb WNT and BMP signaling in human RA-gastruloids and find that these signaling pathways regulate somite patterning and neural tube length, respectively, while genetic perturbation of the transcription factors PAX3 and TBX6 markedly compromises the formation of neural crest and somites/renal cells, respectively. Human RA-gastruloids complement other embryonic organoids in serving as a simple, robust and screenable model for decoding early human embryogenesis.
Collapse
Affiliation(s)
- Nobuhiko Hamazaki
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Wei Yang
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Connor Kubo
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Chengxiang Qiu
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Beth K. Martin
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Riddhiman K. Garge
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - Samuel G. Regalado
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, 98195, USA
| | - Eva Nichols
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Choli Lee
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Riza M. Daza
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Sanjay Srivatsan
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, 98195, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA 98195, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA 98195, USA
| |
Collapse
|
45
|
Ye F, Wang J, Li J, Mei Y, Guo G. Mapping Cell Atlases at the Single-Cell Level. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305449. [PMID: 38145338 PMCID: PMC10885669 DOI: 10.1002/advs.202305449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/01/2023] [Indexed: 12/26/2023]
Abstract
Recent advancements in single-cell technologies have led to rapid developments in the construction of cell atlases. These atlases have the potential to provide detailed information about every cell type in different organisms, enabling the characterization of cellular diversity at the single-cell level. Global efforts in developing comprehensive cell atlases have profound implications for both basic research and clinical applications. This review provides a broad overview of the cellular diversity and dynamics across various biological systems. In addition, the incorporation of machine learning techniques into cell atlas analyses opens up exciting prospects for the field of integrative biology.
Collapse
Affiliation(s)
- Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
- Liangzhu LaboratoryZhejiang UniversityHangzhouZhejiang311121China
| | - Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
- Liangzhu LaboratoryZhejiang UniversityHangzhouZhejiang311121China
| | - Jiaqi Li
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
| | - Yuqing Mei
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
- Liangzhu LaboratoryZhejiang UniversityHangzhouZhejiang311121China
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative MedicineDr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineHangzhouZhejiang310058China
- Institute of HematologyZhejiang UniversityHangzhouZhejiang310000China
| |
Collapse
|
46
|
Yagita K. Emergence of the circadian clock oscillation during the developmental process in mammals. Curr Opin Genet Dev 2024; 84:102152. [PMID: 38266394 DOI: 10.1016/j.gde.2024.102152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 01/26/2024]
Abstract
The circadian clocks are cell-autonomous intrinsic oscillators existing throughout the body to coordinate intracellular and intercellular functions of each organ or tissue. The circadian clock oscillation gradually emerges during mid-to-late gestation in the mammalian developmental process. Recently, it has been revealed that the in vitro differentiation of mouse ES cells recapitulates the circadian clock development. Moreover, reprogramming of the cells results in the redisappearance of the clock, indicating that circadian clocks are tightly coupled with cellular differentiation. Interestingly, before the circadian clock develops, the embryo is governed under ultradian rhythms driven by the segmentation clock. This short review explores these observations, discussing the significance of the emergence of circadian clock oscillation during the mammalian developmental process.
Collapse
Affiliation(s)
- Kazuhiro Yagita
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan.
| |
Collapse
|
47
|
Handford CE, Junyent S, Jorgensen V, Zernicka-Goetz M. Topical section: embryonic models (2023) for Current Opinion in Genetics & Development. Curr Opin Genet Dev 2024; 84:102134. [PMID: 38052116 PMCID: PMC11556421 DOI: 10.1016/j.gde.2023.102134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/20/2023] [Accepted: 11/03/2023] [Indexed: 12/07/2023]
Abstract
Stem cell-based mammalian embryo models facilitate the discovery of developmental mechanisms because they are more amenable to genetic and epigenetic perturbations than natural embryos. Here, we highlight exciting recent advances that have yielded a plethora of models of embryonic development. Imperfections in these models highlight gaps in our current understanding and outline future research directions, ushering in an exciting new era for embryology.
Collapse
Affiliation(s)
- Charlotte E Handford
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA. https://twitter.com/@CEHandford
| | - Sergi Junyent
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA. https://twitter.com/@JunyentSergi
| | - Victoria Jorgensen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Magdalena Zernicka-Goetz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
48
|
Ramesh PS, Chu LF. Species-specific roles of the Notch ligands, receptors, and targets orchestrating the signaling landscape of the segmentation clock. Front Cell Dev Biol 2024; 11:1327227. [PMID: 38348091 PMCID: PMC10859470 DOI: 10.3389/fcell.2023.1327227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/20/2023] [Indexed: 02/15/2024] Open
Abstract
Somitogenesis is a hallmark feature of all vertebrates and some invertebrate species that involves the periodic formation of block-like structures called somites. Somites are transient embryonic segments that eventually establish the entire vertebral column. A highly conserved molecular oscillator called the segmentation clock underlies this periodic event and the pace of this clock regulates the pace of somite formation. Although conserved signaling pathways govern the clock in most vertebrates, the mechanisms underlying the species-specific divergence in various clock characteristics remain elusive. For example, the segmentation clock in classical model species such as zebrafish, chick, and mouse embryos tick with a periodicity of ∼30, ∼90, and ∼120 min respectively. This enables them to form the species-specific number of vertebrae during their overall timespan of somitogenesis. Here, we perform a systematic review of the species-specific features of the segmentation clock with a keen focus on mouse embryos. We perform this review using three different perspectives: Notch-responsive clock genes, ligand-receptor dynamics, and synchronization between neighboring oscillators. We further review reports that use non-classical model organisms and in vitro model systems that complement our current understanding of the segmentation clock. Our review highlights the importance of comparative developmental biology to further our understanding of this essential developmental process.
Collapse
Affiliation(s)
- Pranav S. Ramesh
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Reproductive Biology and Regenerative Medicine Research Group, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Calgary, AB, Canada
| | - Li-Fang Chu
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Reproductive Biology and Regenerative Medicine Research Group, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Calgary, AB, Canada
| |
Collapse
|
49
|
Xu H, Fu H, Long Y, Ang KS, Sethi R, Chong K, Li M, Uddamvathanak R, Lee HK, Ling J, Chen A, Shao L, Liu L, Chen J. Unsupervised spatially embedded deep representation of spatial transcriptomics. Genome Med 2024; 16:12. [PMID: 38217035 PMCID: PMC10790257 DOI: 10.1186/s13073-024-01283-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/02/2024] [Indexed: 01/14/2024] Open
Abstract
Optimal integration of transcriptomics data and associated spatial information is essential towards fully exploiting spatial transcriptomics to dissect tissue heterogeneity and map out inter-cellular communications. We present SEDR, which uses a deep autoencoder coupled with a masked self-supervised learning mechanism to construct a low-dimensional latent representation of gene expression, which is then simultaneously embedded with the corresponding spatial information through a variational graph autoencoder. SEDR achieved higher clustering performance on manually annotated 10 × Visium datasets and better scalability on high-resolution spatial transcriptomics datasets than existing methods. Additionally, we show SEDR's ability to impute and denoise gene expression (URL: https://github.com/JinmiaoChenLab/SEDR/ ).
Collapse
Affiliation(s)
- Hang Xu
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Huazhu Fu
- Institute of High-Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), Singapore, 138632, Singapore
| | - Yahui Long
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Kok Siong Ang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Raman Sethi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Kelvin Chong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Mengwei Li
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Rom Uddamvathanak
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Hong Kai Lee
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Jingjing Ling
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Ao Chen
- BGI Research-Southwest, BGI, Chongqing, 401329, China
- JFL-BGI STOmics Center, Jinfeng Laboratory, Chongqing, 401329, China
| | - Ling Shao
- UCAS-Terminus AI Lab, University of Chinese Academy of Sciences, Beijing, China
| | | | - Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore.
- Immunology Translational Research Program, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), 5 Science Drive 2, BlkMD4, Level 3, Singapore, 117545, Singapore.
| |
Collapse
|
50
|
Shankar V, van Blitterswijk C, Vrij E, Giselbrecht S. Automated, High-Throughput Phenotypic Screening and Analysis Platform to Study Pre- and Post-Implantation Morphogenesis in Stem Cell-Derived Embryo-Like Structures. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304987. [PMID: 37991133 PMCID: PMC10811479 DOI: 10.1002/advs.202304987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/11/2023] [Indexed: 11/23/2023]
Abstract
Combining high-throughput generation and high-content imaging of embryo models will enable large-scale screening assays in the fields of (embryo) toxicity, drug development, embryogenesis, and reproductive medicine. This study shows the continuous culture and in situ (i.e., in microwell) imaging-based readout of a 3D stem cell-based model of peri-implantation epiblast (Epi)/extraembryonic endoderm (XEn) development with an expanded pro-amniotic cavity (PAC) (E3.5 E5.5), namely XEn/EPiCs. Automated image analysis and supervised machine learning permit the identification of embryonic morphogenesis, tissue compartmentalization, cell differentiation, and consecutive classification. Screens with signaling pathway modulators at different time windows provide spatiotemporal information on their phenotypic effect on developmental processes leading to the formation of XEn/EPiCs. Exposure of the biological model in the microwell platform to pathway modulators at two time windows, namely 0-72 h and 48-120 h, show that Wnt and Fgf/MAPK pathway modulators affect Epi differentiation and its polarization, while modulation of BMP and Tgfβ/Nodal pathway affects XEn specification and epithelialization. Further, their collective role is identified in the timing of the formation and expansion of PAC. The newly developed, scalable culture and analysis platform, thereby, provides a unique opportunity to quantitatively and systematically study effects of pathway modulators on early embryonic development.
Collapse
Affiliation(s)
- Vinidhra Shankar
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment for Instructive Biomaterials Engineering (IBE)Maastricht UniversityMaastricht6229ETThe Netherlands
| | - Clemens van Blitterswijk
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment for Instructive Biomaterials Engineering (IBE)Maastricht UniversityMaastricht6229ETThe Netherlands
| | - Erik Vrij
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment for Instructive Biomaterials Engineering (IBE)Maastricht UniversityMaastricht6229ETThe Netherlands
| | - Stefan Giselbrecht
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment for Instructive Biomaterials Engineering (IBE)Maastricht UniversityMaastricht6229ETThe Netherlands
| |
Collapse
|