1
|
Darrigrand JF, Salowka A, Torres-Cano A, Tapia-Rojo R, Zhu T, Garcia-Manyes S, Spagnoli FM. Acinar-ductal cell rearrangement drives branching morphogenesis of the murine pancreas in an IGF/PI3K-dependent manner. Dev Cell 2024; 59:326-338.e5. [PMID: 38237591 PMCID: PMC11805742 DOI: 10.1016/j.devcel.2023.12.011] [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: 01/20/2023] [Revised: 10/24/2023] [Accepted: 12/20/2023] [Indexed: 02/08/2024]
Abstract
During organ formation, progenitor cells need to acquire different cell identities and organize themselves into distinct structural units. How these processes are coordinated and how tissue architecture(s) is preserved despite the dramatic cell rearrangements occurring in developing organs remain unclear. Here, we identified cellular rearrangements between acinar and ductal progenitors as a mechanism to drive branching morphogenesis in the pancreas while preserving the integrity of the acinar-ductal functional unit. Using ex vivo and in vivo mouse models, we found that pancreatic ductal cells form clefts by protruding and pulling on the acinar basement membrane, which leads to acini splitting. Newly formed acini remain connected to the bifurcated branches generated by ductal cell rearrangement. Insulin growth factor (IGF)/phosphatidylinositol 3-kinase (PI3K) pathway finely regulates this process by controlling pancreatic ductal tissue fluidity, with a simultaneous impact on branching and cell fate acquisition. Together, our results explain how acinar structure multiplication and branch bifurcation are synchronized during pancreas organogenesis.
Collapse
Affiliation(s)
- Jean-Francois Darrigrand
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, Great Maze Pond, SE1 9RT London, UK
| | - Anna Salowka
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, Great Maze Pond, SE1 9RT London, UK
| | - Alejo Torres-Cano
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, Great Maze Pond, SE1 9RT London, UK
| | - Rafael Tapia-Rojo
- Department of Physics, London Centre for Nanotechnology, King's College London, London, UK
| | - Tong Zhu
- Department of Physics, Randall Centre for Cell and Molecular Biophysics, Centre for the Physical Science of Life and London Centre for Nanotechnology, King's College London, London, UK; Single-Molecule Mechanobiology Laboratory, The Francis Crick Institute, London, UK
| | - Sergi Garcia-Manyes
- Department of Physics, Randall Centre for Cell and Molecular Biophysics, Centre for the Physical Science of Life and London Centre for Nanotechnology, King's College London, London, UK; Single-Molecule Mechanobiology Laboratory, The Francis Crick Institute, London, UK
| | - Francesca M Spagnoli
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, Great Maze Pond, SE1 9RT London, UK.
| |
Collapse
|
2
|
Agerskov RH, Nyeng P. Innervation of the pancreas in development and disease. Development 2024; 151:dev202254. [PMID: 38265192 DOI: 10.1242/dev.202254] [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: 01/25/2024]
Abstract
The autonomic nervous system innervates the pancreas by sympathetic, parasympathetic and sensory branches during early organogenesis, starting with neural crest cell invasion and formation of an intrinsic neuronal network. Several studies have demonstrated that signals from pancreatic neural crest cells direct pancreatic endocrinogenesis. Likewise, autonomic neurons have been shown to regulate pancreatic islet formation, and have also been implicated in type I diabetes. Here, we provide an overview of recent progress in mapping pancreatic innervation and understanding the interactions between pancreatic neurons, epithelial morphogenesis and cell differentiation. Finally, we discuss pancreas innervation as a factor in the development of diabetes.
Collapse
Affiliation(s)
- Rikke Hoegsberg Agerskov
- Roskilde University, Department of Science and Environment, Universitetsvej 1, building 28, Roskilde 4000, Denmark
| | - Pia Nyeng
- Roskilde University, Department of Science and Environment, Universitetsvej 1, building 28, Roskilde 4000, Denmark
| |
Collapse
|
3
|
Xu X, Seymour PA, Sneppen K, Trusina A, Egeskov-Madsen ALR, Jørgensen MC, Jensen MH, Serup P. Jag1-Notch cis-interaction determines cell fate segregation in pancreatic development. Nat Commun 2023; 14:348. [PMID: 36681690 PMCID: PMC9867774 DOI: 10.1038/s41467-023-35963-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/10/2023] [Indexed: 01/22/2023] Open
Abstract
The Notch ligands Jag1 and Dll1 guide differentiation of multipotent pancreatic progenitor cells (MPCs) into unipotent pro-acinar cells (PACs) and bipotent duct/endocrine progenitors (BPs). Ligand-mediated trans-activation of Notch receptors induces oscillating expression of the transcription factor Hes1, while ligand-receptor cis-interaction indirectly represses Hes1 activation. Despite Dll1 and Jag1 both displaying cis- and trans-interactions, the two mutants have different phenotypes for reasons not fully understood. Here, we present a mathematical model that recapitulates the spatiotemporal differentiation of MPCs into PACs and BPs. The model correctly captures cell fate changes in Notch pathway knockout mice and small molecule inhibitor studies, and a requirement for oscillatory Hes1 expression to maintain the multipotent state. Crucially, the model entails cell-autonomous attenuation of Notch signaling by Jag1-mediated cis-inhibition in MPC differentiation. The model sheds light on the underlying mechanisms, suggesting that cis-interaction is crucial for exiting the multipotent state, while trans-interaction is required for adopting the bipotent fate.
Collapse
Affiliation(s)
- Xiaochan Xu
- The Niels Bohr Institute, University of Copenhagen, DK-2100, Copenhagen Ø, Denmark
| | - Philip Allan Seymour
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, DK-2200, Copenhagen N, Denmark
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, DK-2200, Copenhagen N, Denmark
| | - Kim Sneppen
- The Niels Bohr Institute, University of Copenhagen, DK-2100, Copenhagen Ø, Denmark
| | - Ala Trusina
- The Niels Bohr Institute, University of Copenhagen, DK-2100, Copenhagen Ø, Denmark
| | - Anuska la Rosa Egeskov-Madsen
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, DK-2200, Copenhagen N, Denmark
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, DK-2200, Copenhagen N, Denmark
| | - Mette Christine Jørgensen
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, DK-2200, Copenhagen N, Denmark
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, DK-2200, Copenhagen N, Denmark
| | - Mogens Høgh Jensen
- The Niels Bohr Institute, University of Copenhagen, DK-2100, Copenhagen Ø, Denmark.
| | - Palle Serup
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, DK-2200, Copenhagen N, Denmark.
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, DK-2200, Copenhagen N, Denmark.
| |
Collapse
|
4
|
Freudenblum J, Meyer D, Kimmel RA. Mitochondrial network expansion and dynamic redistribution during islet morphogenesis in zebrafish larvae. FEBS Lett 2023; 597:262-275. [PMID: 36217213 PMCID: PMC10092693 DOI: 10.1002/1873-3468.14508] [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: 06/30/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 01/26/2023]
Abstract
Mitochondria, organelles critical for energy production, modify their shape and location in response to developmental state and metabolic demands. Mitochondria are altered in diabetes, but the mechanistic basis is poorly defined, due to difficulties in assessing mitochondria within an intact organism. Here, we use in vivo imaging in transparent zebrafish larvae to demonstrate filamentous, interconnected mitochondrial networks within islet cells. Mitochondrial movements highly resemble what has been reported for human islet cells in vitro, showing conservation in behaviour across species and cellular context. During islet development, mitochondrial content increases with emergence of cell motility, and mitochondria disperse within fine protrusions. Overall, this work presents quantitative analysis of mitochondria within their native environment and provides insights into mitochondrial behaviour during organogenesis.
Collapse
Affiliation(s)
| | - Dirk Meyer
- Institute of Molecular Biology/CMBIUniversity of InnsbruckAustria
| | - Robin A. Kimmel
- Institute of Molecular Biology/CMBIUniversity of InnsbruckAustria
| |
Collapse
|
5
|
Paramore SV, Goodwin K, Nelson CM. How to build an epithelial tree. Phys Biol 2022; 19. [DOI: 10.1088/1478-3975/ac9e38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 10/27/2022] [Indexed: 11/23/2022]
Abstract
Abstract
Nature has evolved a variety of mechanisms to build epithelial trees of diverse architectures within different organs and across species. Epithelial trees are elaborated through branch initiation and extension, and their morphogenesis ends with branch termination. Each of these steps of the branching process can be driven by the actions of epithelial cells themselves (epithelial-intrinsic mechanisms) or by the cells of their surrounding tissues (epithelial-extrinsic mechanisms). Here, we describe examples of how these mechanisms drive each stage of branching morphogenesis, drawing primarily from studies of the lung, kidney, salivary gland, mammary gland, and pancreas, all of which contain epithelial trees that form through collective cell behaviors. Much of our understanding of epithelial branching comes from experiments using mice, but we also include examples here from avian and reptilian models. Throughout, we highlight how distinct mechanisms are employed in different organs and species to build epithelial trees. We also highlight how similar morphogenetic motifs are used to carry out conserved developmental programs or repurposed to support novel ones. Understanding the unique strategies used by nature to build branched epithelia from across the tree of life can help to inspire creative solutions to problems in tissue engineering and regenerative medicine.
Collapse
|
6
|
Lim R, Banerjee A, Biswas R, Chari AN, Raghavan S. Mechanotransduction through adhesion molecules: Emerging roles in regulating the stem cell niche. Front Cell Dev Biol 2022; 10:966662. [PMID: 36172276 PMCID: PMC9511051 DOI: 10.3389/fcell.2022.966662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
Stem cells have been shown to play an important role in regenerative medicine due to their proliferative and differentiation potential. The challenge, however, lies in regulating and controlling their potential for this purpose. Stem cells are regulated by growth factors as well as an array of biochemical and mechanical signals. While the role of biochemical signals and growth factors in regulating stem cell homeostasis is well explored, the role of mechanical signals has only just started to be investigated. Stem cells interact with their niche or to other stem cells via adhesion molecules that eventually transduce mechanical cues to maintain their homeostatic function. Here, we present a comprehensive review on our current understanding of the influence of the forces perceived by cell adhesion molecules on the regulation of stem cells. Additionally, we provide insights on how this deeper understanding of mechanobiology of stem cells has translated toward therapeutics.
Collapse
Affiliation(s)
- Ryan Lim
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Avinanda Banerjee
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Ritusree Biswas
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
- Sastra University, Thanjavur, TN, India
| | - Anana Nandakumar Chari
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Srikala Raghavan
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
| |
Collapse
|
7
|
Bakhti M, Bastidas-Ponce A, Tritschler S, Czarnecki O, Tarquis-Medina M, Nedvedova E, Jaki J, Willmann SJ, Scheibner K, Cota P, Salinno C, Boldt K, Horn N, Ueffing M, Burtscher I, Theis FJ, Coskun Ü, Lickert H. Synaptotagmin-13 orchestrates pancreatic endocrine cell egression and islet morphogenesis. Nat Commun 2022; 13:4540. [PMID: 35927244 PMCID: PMC9352765 DOI: 10.1038/s41467-022-31862-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
During pancreas development endocrine cells leave the ductal epithelium to form the islets of Langerhans, but the morphogenetic mechanisms are incompletely understood. Here, we identify the Ca2+-independent atypical Synaptotagmin-13 (Syt13) as a key regulator of endocrine cell egression and islet formation. We detect specific upregulation of the Syt13 gene and encoded protein in endocrine precursors and the respective lineage during islet formation. The Syt13 protein is localized to the apical membrane of endocrine precursors and to the front domain of egressing endocrine cells, marking a previously unidentified apical-basal to front-rear repolarization during endocrine precursor cell egression. Knockout of Syt13 impairs endocrine cell egression and skews the α-to-β-cell ratio. Mechanistically, Syt13 is a vesicle trafficking protein, transported via the microtubule cytoskeleton, and interacts with phosphatidylinositol phospholipids for polarized localization. By internalizing a subset of plasma membrane proteins at the front domain, including α6β4 integrins, Syt13 modulates cell-matrix adhesion and allows efficient endocrine cell egression. Altogether, these findings uncover an unexpected role for Syt13 as a morphogenetic driver of endocrinogenesis and islet formation.
Collapse
Affiliation(s)
- Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sophie Tritschler
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Technical University of Munich, School of Life Sciences Weihenstephan, Freising, Germany
| | - Oliver Czarnecki
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Technische Universität München, School of Medicine, München, Germany
| | - Marta Tarquis-Medina
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Technische Universität München, School of Medicine, München, Germany
| | - Eva Nedvedova
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany
- SOTIO a.s, Jankovcova 1518/2, Prague, Czech Republic
| | - Jessica Jaki
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Stefanie J Willmann
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Katharina Scheibner
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Perla Cota
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Technische Universität München, School of Medicine, München, Germany
| | - Ciro Salinno
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Technische Universität München, School of Medicine, München, Germany
| | - Karsten Boldt
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Nicola Horn
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Marius Ueffing
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Technical University of Munich, Department of Mathematics, Garching b, Munich, Germany
| | - Ünal Coskun
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany
- Center of Membrane Biochemistry and Lipid Research, Carl Gustav Carus School of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Technische Universität München, School of Medicine, München, Germany.
| |
Collapse
|
8
|
Liu WW, Hu J, Zhao Y, Wang R, Han Q, Rong XZ, Wang SY, Wang EH, Wu MX, Wang S, Liu Y. PTP-PEST Regulated Membranous/Cytoplasmic Translocation of p120ctn in the Lung Cancer Resistance to Tyrosine Kinase Inhibitor. Appl Immunohistochem Mol Morphol 2022; 30:215-224. [PMID: 35030104 DOI: 10.1097/pai.0000000000001008] [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: 03/16/2021] [Accepted: 12/10/2021] [Indexed: 12/24/2022]
Abstract
Our previous studies indicate that resistance induction using first-generation tyrosine kinase inhibitors (TKIs) in lung cancer is accompanied with p120-catenin (p120ctn) cytoplasmic translocation from the membrane. However, the molecular mechanism underlying p120ctn intracytoplasmic translocation has not yet been reported. We performed immunohistochemistry to detect the correlation of p120ctn distribution with protein tyrosine phosphatase non-receptor type 12 (PTP-PEST) and p120ctn Y335 phosphorylation levels in non-small cell lung cancer (NSCLC) patients. After resistance induction using first-generation TKIs in lung cancer cells, Western blotting and substrate trapping were used to assess PTP-PEST expression and its influence on p120ctn Y335 phosphorylation, as well as the role of p120ctn Y335 phosphorylation on the association of p120ctn with E-cadherin and p120ctn membrane/cytoplasm translocation. In 197 samples collected from NSCLC patients, cytoplasmic p120ctn and enhanced p120ctn Y335 phosphorylation were associated with decreased PTP-PEST. After resistance induction using gefitinib, decreased PTP-PEST expression was accompanied by enhanced phosphorylation of p120ctn Y335 and p120ctn translocated to the cytoplasm. In gefitinib-resistant cells, PTP-PEST overexpression restrained p120ctn Y335 phosphorylation and restored membrane p120ctn expression. PTP-PEST enhanced the interaction of p120ctn with E-cadherin and elevated p120ctn membrane expression. However, increased p120ctn-Y335F mutant had no effect on p120ctn interaction with E-cadherin and membrane/cytoplasm translocation compared with the control group. In conclusion, resistance to first-generation TKIs inhibited PTP-PEST expression, which promoted p120ctn-Y335 phosphorylation and reduced the interaction of p120ctn with E-cadherin, resulting in p120ctn cytoplasmic translocation.
Collapse
Affiliation(s)
- Wei-Wei Liu
- Department of Anesthesiology, The First Hospital of China Medical University
| | - Jing Hu
- Sujia Tuo Town Community Health Service Center, Beijing, PR China
| | - Yue Zhao
- Department of Pathology, The First Hospital of China Medical University and College of Basic Medical Sciences, China Medical University
| | - Rui Wang
- Department of Pathology, The First Hospital of China Medical University and College of Basic Medical Sciences, China Medical University
| | - Qiang Han
- Department of Pathology, The First Hospital of China Medical University and College of Basic Medical Sciences, China Medical University
| | - Xue-Zhu Rong
- Department of Pathology, The First Hospital of China Medical University and College of Basic Medical Sciences, China Medical University
| | - Si-Yao Wang
- Department of Pathology, The First Hospital of China Medical University and College of Basic Medical Sciences, China Medical University
| | - En-Hua Wang
- Department of Pathology, The First Hospital of China Medical University and College of Basic Medical Sciences, China Medical University
| | - Mei-Xi Wu
- China Medical University-The Queen's University of Belfast Joint College, Shenyang
| | - Si Wang
- Department of Medical Microbiology and Human Parasitology, College of Basic Medical Sciences, China Medical University
| | - Yang Liu
- Department of Pathology, The First Hospital of China Medical University and College of Basic Medical Sciences, China Medical University
| |
Collapse
|
9
|
Ahuja N, Cleaver O. The cell cortex as mediator of pancreatic epithelial development and endocrine differentiation. Curr Opin Genet Dev 2022; 72:118-127. [PMID: 34929610 PMCID: PMC8915777 DOI: 10.1016/j.gde.2021.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 02/03/2023]
Abstract
Organogenesis is the complex process of cells coordinating their own proliferation with changes to their shape, cell migration and cell-cell signaling, so that they transform into a three dimensional functional tissue, with its own custom range of differentiated cell types. Understanding when and where critical signals emanate from, and how those signals are transduced and interpreted, is the fundamental challenge of developmental biology. Here, we review recent findings regarding how progenitor cells interpret cues during pancreatic morphogenesis and how they coordinate cell fate determination. Recent evidence suggests that molecules located in the cell cortex play a crticial role in determining cellular behavior during pancreatic morphogenesis. Specifically, we find that control of cell adhesion, polarity, and constriction are all integral to both initiation of epithelial development and to later cell differentiation. Here, we review key molecules that coordinate these processes and suggest that the cell cortex acts as a signaling center that relays cues during pancreas development.
Collapse
Affiliation(s)
- Neha Ahuja
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA.
| |
Collapse
|
10
|
Heilmann S, Semb H, Nyeng P. Quantifying spatial position in a branched structure in immunostained mouse tissue sections. STAR Protoc 2021; 2:100806. [PMID: 34632415 PMCID: PMC8488404 DOI: 10.1016/j.xpro.2021.100806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
We have developed a protocol to quantify the position of a cell in a branched structure based on two-dimensional microscopy images of tissue sections. Biological branched structures include organs such as the lungs, kidneys, and pancreas. In these organs, cell fate has been correlated with position, based on a qualitative estimate. However, a quantitative means of evaluating the cell position has been lacking. With this protocol, the correlation between cell fate and cell position was measured in mouse embryonic pancreas. For complete details on the use and execution of this protocol, please refer to Nyeng et al. (2019).
Collapse
Affiliation(s)
- Silja Heilmann
- Novo Nordisk Foundation Center for Stem Cell Biology (Danstem), University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Henrik Semb
- Novo Nordisk Foundation Center for Stem Cell Biology (Danstem), University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
- Institute of Translational Stem Cell Research, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Pia Nyeng
- Department of Science and Environment, Roskilde University, Universitetsvej 1, Postbox 260, 4000 Roskilde, Denmark
| |
Collapse
|
11
|
Nyeng P, Dela Cruz GV, Semb H. Flow cytometry detection of surface and intracellular antigens in pancreas from a single mouse embryo. STAR Protoc 2021; 2:100636. [PMID: 34258596 PMCID: PMC8255939 DOI: 10.1016/j.xpro.2021.100636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
We here report a flow-cytometry-based protocol to measure single-cell protein expression in small samples. The protocol is optimized for simultaneous detection of fluorescent proteins and intracellular and surface antigens in the embryonic pancreas from the mouse. Owing to low cell numbers, current protocols for flow cytometric analysis of embryonic tissues rely on tissue pooling. Our protocol enables analysis of one pancreas per sample, thereby facilitating detection of biological variation and minimizing the number of experimental animals needed. For complete details on the use and execution of this protocol, please refer to Nyeng et al (2019). Quantitative protein analysis of a single embryonic pancreas using flow cytometry Simultaneous detection of reporter, cell surface, and intracellular proteins Enables detection of biological variation and reduces the number of animals needed Enables the study of heterogeneous cellular protein expression during organogenesis
Collapse
Affiliation(s)
- Pia Nyeng
- Department of Science and Environment, Roskilde University, Universitetsvej 1, Postbox 260, DK-4000 Roskilde, Denmark
| | - Gelo Victoriano Dela Cruz
- Novo Nordisk Foundation Center for Stem Cell Biology (Danstem), University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen N, Denmark
| | - Henrik Semb
- Novo Nordisk Foundation Center for Stem Cell Biology (Danstem), University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen N, Denmark.,Institute of Translational Stem Cell Research, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| |
Collapse
|
12
|
Isaacson A, Spagnoli FM. Pancreatic cell fate specification: insights into developmental mechanisms and their application for lineage reprogramming. Curr Opin Genet Dev 2021; 70:32-39. [PMID: 34062490 DOI: 10.1016/j.gde.2021.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/01/2021] [Accepted: 05/02/2021] [Indexed: 12/15/2022]
Abstract
Diabetes is a group of metabolic disorders, which results from insufficient functional pancreatic β-cell mass either due to the autoimmune destruction of insulin producing β-cells, or their death or de-differentiation as compensation for insulin resistance. The ability to reprogram cell types within close developmental proximity to β-cells offers a strategy to replenish β-cell mass and a future possible treatment of diabetes. Here, we review recent advances in the fields of pancreas development and lineage reprogramming. We also probe the possibility of using reprogrammed cells as an approach by which to further understand developmental mechanisms, in particular roadblocks to changing cell identity. Finally, we highlight fundamental challenges that need to be overcome to advance lineage reprogramming for generating pancreatic cells.
Collapse
Affiliation(s)
- Abigail Isaacson
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London SE1 9RT, UK
| | - Francesca M Spagnoli
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London SE1 9RT, UK.
| |
Collapse
|
13
|
Zhang X, Ma Z, Song E, Xu T. Islet organoid as a promising model for diabetes. Protein Cell 2021; 13:239-257. [PMID: 33751396 PMCID: PMC7943334 DOI: 10.1007/s13238-021-00831-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
Studies on diabetes have long been hampered by a lack of authentic disease models that, ideally, should be unlimited and able to recapitulate the abnormalities involved in the development, structure, and function of human pancreatic islets under pathological conditions. Stem cell-based islet organoids faithfully recapitulate islet development in vitro and provide large amounts of three-dimensional functional islet biomimetic materials with a morphological structure and cellular composition similar to those of native islets. Thus, islet organoids hold great promise for modeling islet development and function, deciphering the mechanisms underlying the onset of diabetes, providing an in vitro human organ model for infection of viruses such as SARS-CoV-2, and contributing to drug screening and autologous islet transplantation. However, the currently established islet organoids are generally immature compared with native islets, and further efforts should be made to improve the heterogeneity and functionality of islet organoids, making it an authentic and informative disease model for diabetes. Here, we review the advances and challenges in the generation of islet organoids, focusing on human pluripotent stem cell-derived islet organoids, and the potential applications of islet organoids as disease models and regenerative therapies for diabetes.
Collapse
Affiliation(s)
- Xiaofei Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhuo Ma
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Eli Song
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China. .,Guangzhou Regenerative Medicine and Health Guangdong Laboratory (Bioland Laboratory), Guangzhou, 510005, China.
| |
Collapse
|
14
|
Lee SN, Kim SJ, Yoon SA, Song JM, Ahn JS, Kim HC, Choi AMK, Yoon JH. CD44v3-Positive Intermediate Progenitor Cells Contribute to Airway Goblet Cell Hyperplasia. Am J Respir Cell Mol Biol 2021; 64:247-259. [PMID: 33264080 DOI: 10.1165/rcmb.2020-0350oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022] Open
Abstract
In allergic airway diseases, intermediate progenitor cells (IPCs) increase in number in the surface epithelium. IPCs arise from basal cells, the origin of hallmark pathological changes, including goblet cell hyperplasia and mucus hypersecretion. Thus, targeting IPCs will benefit future treatment of allergic airway diseases. However, the lack of adequate cell surface markers for IPCs limits their identification and characterization. We now show that CD44 containing exon v3 (CD44v3) is a surface marker for IPCs that are capable of both proliferating and generating differentiated goblet cells in allergic human nasal epithelium. In primary human nasal epithelial cells that had differentiated at an air-liquid interface, IL-4 upregulated mRNA expression of three CD44v variants that include exon v3 (CD44v3-v6, CD44v3,v8-v10, and CD44v3-v10), and it induced expression of CD44v3 protein in the basal and suprabasal layers of the culture. FACS analysis revealed two subpopulations differing in CD44v3 concentrations, as follows: CD44v3low cells expressed high amounts of proliferative and basal cell markers (Ki-67 and TP63), whereas CD44v3high cells strongly expressed progenitor and immature and mature goblet cell markers (SOX2, CA2, and SPDEF). Importantly, a blocking anti-CD44 antibody suppressed IL-4-induced mucin production by human nasal epithelial cells. Furthermore, CD44v3 was coexpressed with TP63, KRT5, or SOX2 and was upregulated in the basal and suprabasal layers of the nasal surface epithelium of subjects with allergic rhinitis. Taken together, these data demonstrate that high CD44v3 expression contributes to goblet cell hyperplasia in inflammation of the allergic airway.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Augustine M K Choi
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, New York; and.,Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, New York
| | - Joo-Heon Yoon
- The Airway Mucus Institute and.,Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
15
|
Abstract
The pancreas of adult mammals displays a branched structure which transports digestive enzymes produced in the distal acini through a tree-like network of ducts into the duodenum. In contrast to several other branched organs, its branching patterns are not stereotypic. Moreover, the branches do not grow from dichotomic splitting of an initial stem but rather from the formation of microlumen in a mass of cells. These lumen progressively assemble into a hyperconnected network that refines into a tree by the time of birth. We review the cell remodeling events and the molecular mechanisms governing pancreas branching, as well as the role of the surrounding tissues in this process. Furthermore, we draw parallels with other branched organs such as the salivary and mammary gland.
Collapse
Affiliation(s)
- Lydie Flasse
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| | - Coline Schewin
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Anne Grapin-Botton
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany; The Novo Nordisk Foundation Center for Stem Cell Biology, Copenhagen, Denmark.
| |
Collapse
|
16
|
Freudenblum J, Meyer D, Kimmel RA. Inducible Mosaic Cell Labeling Provides Insights Into Pancreatic Islet Morphogenesis. Front Cell Dev Biol 2020; 8:586651. [PMID: 33102488 PMCID: PMC7546031 DOI: 10.3389/fcell.2020.586651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/02/2020] [Indexed: 11/13/2022] Open
Abstract
Pancreatic islets, discrete microorgans embedded within the exocrine pancreas, contain beta cells which are critical for glucose homeostasis. Loss or dysfunction of beta cells leads to diabetes, a disease with expanding global prevalence, and for which regenerative therapies are actively being pursued. Recent efforts have focused on producing mature beta cells in vitro, but it is increasingly recognized that achieving a faithful three-dimensional islet structure is crucial for generating fully functional beta cells. Our current understanding of islet morphogenesis is far from complete, due to the deep internal location of the pancreas in mammalian models, which hampers direct visualization. Zebrafish is a model system well suited for studies of pancreas morphogenesis due to its transparency and the accessible location of the larval pancreas. In order to further clarify the cellular mechanisms of islet formation, we have developed new tools for in vivo visualization of single-cell dynamics. Our results show that clustering islet cells make contact and interconnect through dynamic actin-rich processes, move together while remaining in close proximity to the duct, and maintain high protrusive motility after forming clusters. Quantitative analyses of cell morphology and motility in 3-dimensions lays the groundwork to define therapeutically applicable factors responsible for orchestrating the morphogenic behaviors of coalescing endocrine cells.
Collapse
Affiliation(s)
- Julia Freudenblum
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| | - Dirk Meyer
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| | - Robin A Kimmel
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
17
|
Tracing the cellular basis of islet specification in mouse pancreas. Nat Commun 2020; 11:5037. [PMID: 33028844 PMCID: PMC7541446 DOI: 10.1038/s41467-020-18837-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic islets play an essential role in regulating blood glucose level. Although the molecular pathways underlying islet cell differentiation are beginning to be resolved, the cellular basis of islet morphogenesis and fate allocation remain unclear. By combining unbiased and targeted lineage tracing, we address the events leading to islet formation in the mouse. From the statistical analysis of clones induced at multiple embryonic timepoints, here we show that, during the secondary transition, islet formation involves the aggregation of multiple equipotent endocrine progenitors that transition from a phase of stochastic amplification by cell division into a phase of sublineage restriction and limited islet fission. Together, these results explain quantitatively the heterogeneous size distribution and degree of polyclonality of maturing islets, as well as dispersion of progenitors within and between islets. Further, our results show that, during the secondary transition, α- and β-cells are generated in a contemporary manner. Together, these findings provide insight into the cellular basis of islet development. The cellular basis of islet morphogenesis and fate allocation remain unclear. Here, the authors use a R26-CreER-R26R-Confetti mouse line to follow quantitatively the clonal dynamics of islet formation showing how, during the secondary transition, islet progenitors amplify through rounds of stochastic cell division before becoming restricted to α and β cell sublineages.
Collapse
|
18
|
Abstract
The existence of progenitors within pancreatic ducts has been studied for decades, but the hypothesis that they may help regenerate the adult endocrine compartment (chiefly insulin-producing β-cells) remains contentious. Here, we examine the single-cell transcriptome of the human ductal tree. Our data confirm the paradigm-shifting notion that specific lineages, long thought to be cast in stone, are in fact in a state of flux between differentiation stages. In addition to pro-ductal and pro-acinar transcriptomic gradients, our analysis suggests the existence of a third (ducto-endocrine) differentiation axis. Such prediction was experimentally validated by transplanting sorted progenitor-like cells, which revealed their tri-lineage differentiation potential. Our findings further indicate that progenitors might be activated in situ for therapeutic purposes. We have described multipotent progenitor-like cells within the major pancreatic ducts (MPDs) of the human pancreas. They express PDX1, its surrogate surface marker P2RY1, and the bone morphogenetic protein (BMP) receptor 1A (BMPR1A)/activin-like kinase 3 (ALK3), but not carbonic anhydrase II (CAII). Here we report the single-cell RNA sequencing (scRNA-seq) of ALK3bright+-sorted ductal cells, a fraction that harbors BMP-responsive progenitor-like cells. Our analysis unveiled the existence of multiple subpopulations along two major axes, one that encompasses a gradient of ductal cell differentiation stages, and another featuring cells with transitional phenotypes toward acinar tissue. A third potential ducto-endocrine axis is revealed upon integration of the ALK3bright+ dataset with a single-cell whole-pancreas transcriptome. When transplanted into immunodeficient mice, P2RY1+/ALK3bright+ populations (enriched in PDX1+/ALK3+/CAII− cells) differentiate into all pancreatic lineages, including functional β-cells. This process is accelerated when hosts are treated systemically with an ALK3 agonist. We found PDX1+/ALK3+/CAII− progenitor-like cells in the MPDs of types 1 and 2 diabetes donors, regardless of the duration of the disease. Our findings open the door to the pharmacological activation of progenitor cells in situ.
Collapse
|
19
|
Venhuizen JH, Jacobs FJ, Span PN, Zegers MM. P120 and E-cadherin: Double-edged swords in tumor metastasis. Semin Cancer Biol 2020; 60:107-120. [DOI: 10.1016/j.semcancer.2019.07.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022]
|
20
|
Bakhti M, Scheibner K, Tritschler S, Bastidas-Ponce A, Tarquis-Medina M, Theis FJ, Lickert H. Establishment of a high-resolution 3D modeling system for studying pancreatic epithelial cell biology in vitro. Mol Metab 2019; 30:16-29. [PMID: 31767167 PMCID: PMC6812400 DOI: 10.1016/j.molmet.2019.09.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/06/2019] [Accepted: 09/12/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Translation of basic research from bench-to-bedside relies on a better understanding of similarities and differences between mouse and human cell biology, tissue formation, and organogenesis. Thus, establishing ex vivo modeling systems of mouse and human pancreas development will help not only to understand evolutionary conserved mechanisms of differentiation and morphogenesis but also to understand pathomechanisms of disease and design strategies for tissue engineering. METHODS Here, we established a simple and reproducible Matrigel-based three-dimensional (3D) cyst culture model system of mouse and human pancreatic progenitors (PPs) to study pancreatic epithelialization and endocrinogenesis ex vivo. In addition, we reanalyzed previously reported single-cell RNA sequencing (scRNA-seq) of mouse and human pancreatic lineages to obtain a comprehensive picture of differential expression of key transcription factors (TFs), cell-cell adhesion molecules and cell polarity components in PPs during endocrinogenesis. RESULTS We generated mouse and human polarized pancreatic epithelial cysts derived from PPs. This system allowed to monitor establishment of pancreatic epithelial polarity and lumen formation in cellular and sub-cellular resolution in a dynamic time-resolved fashion. Furthermore, both mouse and human pancreatic cysts were able to differentiate towards the endocrine fate. This differentiation system together with scRNA-seq analysis revealed how apical-basal polarity and tight and adherens junctions change during endocrine differentiation. CONCLUSIONS We have established a simple 3D pancreatic cyst culture system that allows to tempo-spatial resolve cellular and subcellular processes on the mechanistical level, which is otherwise not possible in vivo.
Collapse
Affiliation(s)
- Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), D-85764, Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764, Neuherberg, Germany.
| | - Katharina Scheibner
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), D-85764, Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, Munich, Germany
| | - Sophie Tritschler
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764, Neuherberg, Germany; Institute of Computational Biology, Helmholtz Zentrum München, D-85764, Neuherberg, Germany; Technical University of Munich, School of Life Sciences Weihenstephan, Freising, Germany
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), D-85764, Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, Munich, Germany
| | - Marta Tarquis-Medina
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), D-85764, Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, Munich, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, D-85764, Neuherberg, Germany; Technical University of Munich, Department of Mathematics, Munich, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), D-85764, Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, Munich, Germany.
| |
Collapse
|
21
|
Mariniello K, Ruiz-Babot G, McGaugh EC, Nicholson JG, Gualtieri A, Gaston-Massuet C, Nostro MC, Guasti L. Stem Cells, Self-Renewal, and Lineage Commitment in the Endocrine System. Front Endocrinol (Lausanne) 2019; 10:772. [PMID: 31781041 PMCID: PMC6856655 DOI: 10.3389/fendo.2019.00772] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022] Open
Abstract
The endocrine system coordinates a wide array of body functions mainly through secretion of hormones and their actions on target tissues. Over the last decades, a collective effort between developmental biologists, geneticists, and stem cell biologists has generated a wealth of knowledge related to the contribution of stem/progenitor cells to both organogenesis and self-renewal of endocrine organs. This review provides an up-to-date and comprehensive overview of the role of tissue stem cells in the development and self-renewal of endocrine organs. Pathways governing crucial steps in both development and stemness maintenance, and that are known to be frequently altered in a wide array of endocrine disorders, including cancer, are also described. Crucially, this plethora of information is being channeled into the development of potential new cell-based treatment modalities for endocrine-related illnesses, some of which have made it through clinical trials.
Collapse
Affiliation(s)
- Katia Mariniello
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gerard Ruiz-Babot
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
| | - Emily C. McGaugh
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - James G. Nicholson
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Angelica Gualtieri
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Maria Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
22
|
Abstract
How organ morphogenesis specifies cell fate and whether organ progenitors are predetermined or specified via niche signals are critical developmental biology questions. In this issue of Developmental Cell, Nyeng et al. (2019) modulate cell-cell adhesion in the pancreas and provide evidence that progenitors are plastic and instructed by niche signals.
Collapse
Affiliation(s)
- Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Technical University of Munich, Medical Faculty, Munich, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Technical University of Munich, Medical Faculty, Munich, Germany.
| |
Collapse
|
23
|
Scheibner K, Bakhti M, Bastidas-Ponce A, Lickert H. Wnt signaling: implications in endoderm development and pancreas organogenesis. Curr Opin Cell Biol 2019; 61:48-55. [PMID: 31377680 DOI: 10.1016/j.ceb.2019.07.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023]
Abstract
The pancreas is derived from the foregut endoderm during embryonic development. After gastrulation and endoderm germ layer formation complex morphogenetic events coupled with cell differentiation programs pattern the gut tube and induce pancreas organogenesis. This results in formation of exocrine, ductal and hormone-producing endocrine cells. Among these, endocrine cells are responsible for blood glucose homeostasis and their malfunction leads to diabetes mellitus, which cannot be stopped or reversed by the current standard treatments. Thus, intense efforts to regenerate or replace the lost or dysfunctional insulin-producing β-cells are on the way. This depends on identifying the factors that coordinate pancreas organogenesis. Here, we highlight the contribution of canonical and non-canonical Wnt signaling branches in orchestrating endoderm formation, pancreatic morphogenesis as well as endocrine cell formation and function.
Collapse
Affiliation(s)
- Katharina Scheibner
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany; Technical University of Munich, School of Medicine, Munich, Germany
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany; Technical University of Munich, School of Medicine, Munich, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany; Technical University of Munich, School of Medicine, Munich, Germany.
| |
Collapse
|
24
|
Bastidas-Ponce A, Tritschler S, Dony L, Scheibner K, Tarquis-Medina M, Salinno C, Schirge S, Burtscher I, Böttcher A, Theis F, Lickert H, Bakhti M. Massive single-cell mRNA profiling reveals a detailed roadmap for pancreatic endocrinogenesis. Development 2019; 146:dev.173849. [DOI: 10.1242/dev.173849] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 05/21/2019] [Indexed: 12/21/2022]
Abstract
Deciphering mechanisms of endocrine cell induction, specification and lineage allocation in vivo will provide valuable insights into how the islets of Langerhans are generated. Currently, it is ill defined how endocrine progenitors segregate into different endocrine subtypes during development. Here, we generated a novel Neurogenin3 (Ngn3)-Venus fusion (NVF) reporter mouse line, that closely mirrors the transient endogenous Ngn3 protein expression. To define an in vivo roadmap of endocrinogenesis, we performed single-cell RNA-sequencing of 36,351 pancreatic epithelial and NVF+ cells during secondary transition. This allowed to time-resolve and distinguish Ngn3low endocrine progenitors, Ngn3high endocrine precursors, Fev+ endocrine lineage and hormone+ endocrine subtypes and delineate molecular programs during the stepwise lineage restriction steps. Strikingly, we identified 58 novel signature genes that show the same transient expression dynamics as Ngn3 in the 7,260 profiled Ngn3-expressing cells. The differential expression of these genes in endocrine precursors associated with their cell-fate allocation towards distinct endocrine cell types. Thus, the generation of an accurately regulated NVF reporter allowed us to temporally resolve endocrine lineage development to provide a fine-grained single-cell molecular profile of endocrinogenesis in vivo.
Collapse
Affiliation(s)
- Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- Technical University of Munich, School of Medicine, Munich, Germany
| | - Sophie Tritschler
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- Technical University of Munich, School of Life Sciences Weihenstephan, Freising, Germany
| | - Leander Dony
- Institute of Computational Biology, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany
| | - Katharina Scheibner
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- Technical University of Munich, School of Medicine, Munich, Germany
| | - Marta Tarquis-Medina
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- Technical University of Munich, School of Medicine, Munich, Germany
| | - Ciro Salinno
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- Technical University of Munich, School of Medicine, Munich, Germany
| | - Silvia Schirge
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
| | - Anika Böttcher
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
| | - Fabian Theis
- Institute of Computational Biology, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- Technical University of Munich, Department of Mathematics, Munich, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- Technical University of Munich, School of Medicine, Munich, Germany
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
| |
Collapse
|