1
|
Villalba A, Gitton Y, Inoue M, Aiello V, Blain R, Toupin M, Mazaud-Guittot S, Rachdi L, Semb H, Chédotal A, Scharfmann R. A 3D atlas of the human developing pancreas to explore progenitor proliferation and differentiation. Diabetologia 2024; 67:1066-1078. [PMID: 38630142 PMCID: PMC11058870 DOI: 10.1007/s00125-024-06143-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/07/2024] [Indexed: 04/30/2024]
Abstract
AIMS/HYPOTHESIS Rodent pancreas development has been described in great detail. On the other hand, there are still gaps in our understanding of the developmental trajectories of pancreatic cells during human ontogenesis. Here, our aim was to map the spatial and chronological dynamics of human pancreatic cell differentiation and proliferation by using 3D imaging of cleared human embryonic and fetal pancreases. METHODS We combined tissue clearing with light-sheet fluorescence imaging in human embryonic and fetal pancreases during the first trimester of pregnancy. In addition, we validated an explant culture system enabling in vitro proliferation of pancreatic progenitors to determine the mitogenic effect of candidate molecules. RESULTS We detected the first insulin-positive cells as early as five post-conceptional weeks, two weeks earlier than previously observed. We observed few insulin-positive clusters at five post-conceptional weeks (mean ± SD 9.25±5.65) with a sharp increase to 11 post-conceptional weeks (4307±152.34). We identified a central niche as the location of onset of the earliest insulin cell production and detected extra-pancreatic loci within the adjacent developing gut. Conversely, proliferating pancreatic progenitors were located in the periphery of the epithelium, suggesting the existence of two separated pancreatic niches for differentiation and proliferation. Additionally, we observed that the proliferation ratio of progenitors ranged between 20% and 30%, while for insulin-positive cells it was 1%. We next unveiled a mitogenic effect of the platelet-derived growth factor AA isoform (PDGFAA) in progenitors acting through the pancreatic mesenchyme by increasing threefold the number of proliferating progenitors. CONCLUSIONS/INTERPRETATION This work presents a first 3D atlas of the human developing pancreas, charting both endocrine and proliferating cells across early development.
Collapse
Affiliation(s)
- Adrian Villalba
- Institut Cochin, CNRS, Inserm, Université Paris Cité, Paris, France
| | - Yorick Gitton
- Inserm, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Megumi Inoue
- Inserm, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Virginie Aiello
- Institut Cochin, CNRS, Inserm, Université Paris Cité, Paris, France
| | - Raphaël Blain
- Inserm, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Maryne Toupin
- Inserm, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, Université Rennes, Rennes, France
| | - Séverine Mazaud-Guittot
- Inserm, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, Université Rennes, Rennes, France
| | - Latif Rachdi
- Institut Cochin, CNRS, Inserm, Université Paris Cité, Paris, France
| | - Henrik Semb
- Institute of Translational Stem Cell Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, München, Germany
| | - Alain Chédotal
- Inserm, CNRS, Institut de la Vision, Sorbonne Université, Paris, France.
- Institut de pathologie, groupe hospitalier Est, hospices civils de Lyon, Lyon, France.
- MeLiS, CNRS UMR5284, Inserm U1314, University Claude Bernard Lyon 1, Lyon, France.
| | | |
Collapse
|
2
|
Mubarak HA, Kamal MM, Mahmoud Y, Abd-Elsamea FS, Abdelbary E, Gamea MG, El-Mahdy RI. The ameliorating effects of mesenchymal stem cells compared to α-tocopherol on apoptosis and autophagy in streptozotocin-induced diabetic rats: Implication of PI3K/Akt signaling pathway and entero-insular axis. J Cell Biochem 2023; 124:1705-1719. [PMID: 37796145 DOI: 10.1002/jcb.30482] [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: 04/03/2023] [Revised: 07/21/2023] [Accepted: 09/17/2023] [Indexed: 10/06/2023]
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs) are considered a novel regenerative therapy that holds much potential. This study aimed to examine and compare the ameliorative effects of BM-MSCs compared to α-tocopherol (α-Toc) on apoptosis, autophagy, and β-cell function in a rat model of streptozotocin (STZ)-induced diabetes and further analyzed the implications and interrelations of the entero-insular axis, and type I phosphoinositide 3-kinase (PI3K)/Akt signaling. Forty adult male albino rats were categorized into four groups (n = 10, in each): control group, STZ-induced diabetic group (single i.p. injection of STZ 45 mg/kg), diabetic and treated with BM-MSCs injection, diabetic and treatment with α-Toc p.o. The serum glucose, insulin, nitric oxide (NO), and catalase (CAT) were measured. Histopathological examination of the pancreas, the expression levels of insulin, CD44, caspase-3, autophagy markers, P13K/Akt, and pancreas/duodenum homeobox protein 1, in pancreatic tissue, and glucose-dependent insulinotropic polypeptide (GIP) in the duodenum were detected by hematoxylin and eosin staining, immunofluorescence labeling, and by quantitative real-time polymerase chain reaction. The diabetic rats showed reduced insulin, hyperglycemia, nitrosative stress (NO, CAT), augmented apoptosis (caspase 3), impaired autophagy (p62/SQSTM1, LC3), downregulated PI3K/Akt pathway and increased GIP expression, and degeneration of pancreatic islets. Treatment with either BM-MSCs or α-Toc suppressed the nitrosative stress, reduced apoptosis, recovered autophagy, upregulated PI3K/Akt pathway, and subsequently increased insulin levels, decreased blood glucose, and downregulated GIP expression with partial restoration of pancreatic islets. Based on our findings, the cytoprotective effects of BM-MSCs and α-Toc in type 1-induced diabetes appeared to be related to repaired autophagy and recovered PI3K/Akt signaling. Moreover, we reported their novel effects on reversing intestinal GIP expression level. The effect of BM-MSCs was notably superior to that of α-Toc.
Collapse
Affiliation(s)
- Heba A Mubarak
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Manal M Kamal
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Yossra Mahmoud
- Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Fatma S Abd-Elsamea
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Eman Abdelbary
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Marwa G Gamea
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Reham I El-Mahdy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
- Department of Biochemistry and Physiology, West of Assiut, New Naser City, Badr University, Assiut, Egypt
| |
Collapse
|
3
|
Identification and implication of tissue-enriched ligands in epithelial-endothelial crosstalk during pancreas development. Sci Rep 2022; 12:12498. [PMID: 35864120 PMCID: PMC9304391 DOI: 10.1038/s41598-022-16072-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/04/2022] [Indexed: 11/17/2022] Open
Abstract
Development of the pancreas is driven by an intrinsic program coordinated with signals from other cell types in the epithelial environment. These intercellular communications have been so far challenging to study because of the low concentration, localized production and diversity of the signals released. Here, we combined scRNAseq data with a computational interactomic approach to identify signals involved in the reciprocal interactions between the various cell types of the developing pancreas. This in silico approach yielded 40,607 potential ligand-target interactions between the different main pancreatic cell types. Among this vast network of interactions, we focused on three ligands potentially involved in communications between epithelial and endothelial cells. BMP7 and WNT7B, expressed by pancreatic epithelial cells and predicted to target endothelial cells, and SEMA6D, involved in the reverse interaction. In situ hybridization confirmed the localized expression of Bmp7 in the pancreatic epithelial tip cells and of Wnt7b in the trunk cells. On the contrary, Sema6d was enriched in endothelial cells. Functional experiments on ex vivo cultured pancreatic explants indicated that tip cell-produced BMP7 limited development of endothelial cells. This work identified ligands with a restricted tissular and cellular distribution and highlighted the role of BMP7 in the intercellular communications contributing to vessel development and organization during pancreas organogenesis.
Collapse
|
4
|
Glorieux L, Sapala A, Willnow D, Moulis M, Salowka A, Darrigrand JF, Edri S, Schonblum A, Sakhneny L, Schaumann L, Gómez HF, Lang C, Conrad L, Guillemot F, Levenberg S, Landsman L, Iber D, Pierreux CE, Spagnoli FM. Development of a 3D atlas of the embryonic pancreas for topological and quantitative analysis of heterologous cell interactions. Development 2022; 149:274013. [PMID: 35037942 PMCID: PMC8918780 DOI: 10.1242/dev.199655] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 12/20/2021] [Indexed: 01/05/2023]
Abstract
Generating comprehensive image maps, while preserving spatial three-dimensional (3D) context, is essential in order to locate and assess quantitatively specific cellular features and cell-cell interactions during organ development. Despite recent advances in 3D imaging approaches, our current knowledge of the spatial organization of distinct cell types in the embryonic pancreatic tissue is still largely based on two-dimensional histological sections. Here, we present a light-sheet fluorescence microscopy approach to image the pancreas in three dimensions and map tissue interactions at key time points in the mouse embryo. We demonstrate the utility of the approach by providing volumetric data, 3D distribution of three main cellular components (epithelial, mesenchymal and endothelial cells) within the developing pancreas, and quantification of their relative cellular abundance within the tissue. Interestingly, our 3D images show that endocrine cells are constantly and increasingly in contact with endothelial cells forming small vessels, whereas the interactions with mesenchymal cells decrease over time. These findings suggest distinct cell-cell interaction requirements for early endocrine cell specification and late differentiation. Lastly, we combine our image data in an open-source online repository (referred to as the Pancreas Embryonic Cell Atlas). Summary: A light-sheet fluorescence microscopy approach is used for 3D imaging of the pancreas and to quantitatively map its interactions with surrounding tissues at key development time points in the mouse embryo.
Collapse
Affiliation(s)
- Laura Glorieux
- Cell Biology Unit, de Duve Institute, UCLouvain, Woluwe 1200, Belgium
| | - Aleksandra Sapala
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | - David Willnow
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Manon Moulis
- Cell Biology Unit, de Duve Institute, UCLouvain, Woluwe 1200, Belgium
| | - Anna Salowka
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Jean-Francois Darrigrand
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Shlomit Edri
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Anat Schonblum
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Lina Sakhneny
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Laura Schaumann
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | - Harold F Gómez
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | - Christine Lang
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | - Lisa Conrad
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | | | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Limor Landsman
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dagmar Iber
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | | | - Francesca M Spagnoli
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| |
Collapse
|
5
|
Ghezelayagh Z, Zabihi M, Kazemi Ashtiani M, Ghezelayagh Z, Lynn FC, Tahamtani Y. Recapitulating pancreatic cell-cell interactions through bioengineering approaches: the momentous role of non-epithelial cells for diabetes cell therapy. Cell Mol Life Sci 2021; 78:7107-7132. [PMID: 34613423 PMCID: PMC11072828 DOI: 10.1007/s00018-021-03951-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/09/2021] [Accepted: 09/23/2021] [Indexed: 12/11/2022]
Abstract
Over the past few years, extensive efforts have been made to generate in-vitro pancreatic micro-tissue, for disease modeling or cell replacement approaches in pancreatic related diseases such as diabetes mellitus. To obtain these goals, a closer look at the diverse cells participating in pancreatic development is necessary. Five major non-epithelial pancreatic (pN-Epi) cell populations namely, pancreatic endothelium, mesothelium, neural crests, pericytes, and stellate cells exist in pancreas throughout its development, and they are hypothesized to be endogenous inducers of the development. In this review, we discuss different pN-Epi cells migrating to and existing within the pancreas and their diverse effects on pancreatic epithelium during organ development mediated via associated signaling pathways, soluble factors or mechanical cell-cell interactions. In-vivo and in-vitro experiments, with a focus on N-Epi cells' impact on pancreas endocrine development, have also been considered. Pluripotent stem cell technology and multicellular three-dimensional organoids as new approaches to generate pancreatic micro-tissues have also been discussed. Main challenges for reaching a detailed understanding of the role of pN-Epi cells in pancreas development in utilizing for in-vitro recapitulation have been summarized. Finally, various novel and innovative large-scale bioengineering approaches which may help to recapitulate cell-cell interactions and are crucial for generation of large-scale in-vitro multicellular pancreatic micro-tissues, are discussed.
Collapse
Affiliation(s)
- Zahra Ghezelayagh
- Department of Developmental Biology, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Zabihi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Genetics, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Mohammad Kazemi Ashtiani
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zeinab Ghezelayagh
- Department of Developmental Biology, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, ACECR, Tehran, Iran
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Francis C Lynn
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Surgery and School of Biomedical Engineering , University of British Columbia, Vancouver, BC, Canada
| | - Yaser Tahamtani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
6
|
Ghezelayagh Z, Zabihi M, Zarkesh I, Gonçalves CAC, Larsen M, Hagh-Parast N, Pakzad M, Vosough M, Arjmand B, Baharvand H, Larijani B, Grapin-Botton A, Aghayan HR, Tahamtani Y. Improved Differentiation of hESC-Derived Pancreatic Progenitors by Using Human Fetal Pancreatic Mesenchymal Cells in a Micro-scalable Three-Dimensional Co-culture System. Stem Cell Rev Rep 2021; 18:360-377. [PMID: 34586606 DOI: 10.1007/s12015-021-10266-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2021] [Indexed: 01/12/2023]
Abstract
Mesenchymal cells of diverse origins differ in gene and protein expression besides producing varying effects on their organ-matched epithelial cells' maintenance and differentiation capacity. Co-culture with rodent's tissue-specific pancreatic mesenchyme accelerates proliferation, self-renewal, and differentiation of pancreatic epithelial progenitors. Therefore, in our study, the impact of three-dimensional (3D) co-culture of human fetal pancreatic-derived mesenchymal cells (hFP-MCs) with human embryonic stem cell-derived pancreatic progenitors (hESC-PPs) development towards endocrine and beta cells was assessed. Besides, the ability to maintain scalable cultures combining hFP-MCs and hESC-PPs was investigated. hFP-MCs expressed many markers in common with bone marrow-derived mesenchymal stem cells (BM-MSCs). However, they showed higher expression of DESMIN compared to BM-MSCs. After co-culture of hESC-PPs with hFP-MCs, the pancreatic progenitor (PP) spheroids generated in Matrigel had higher expression of NGN3 and INSULIN than BM-MSCs co-culture group, which shows an inductive impact of pancreatic mesenchyme on hESC-PPs beta-cells maturation. Pancreatic aggregates generated by forced aggregation through scalable AggreWell system showed similar features compared to the spheroids. These aggregates, a combination of hFP-MCs and hESC-PPs, can be applied as an appropriate tool for assessing endocrine-niche interactions and developmental processes by mimicking the pancreatic tissue.
Collapse
Affiliation(s)
- Zahra Ghezelayagh
- Department of Developmental Biology, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, ACECR, Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Zabihi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Genetics, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Ibrahim Zarkesh
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Carla A C Gonçalves
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Larsen
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Newsha Hagh-Parast
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Pakzad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Baharvand
- Department of Developmental Biology, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, ACECR, Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Anne Grapin-Botton
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Yaser Tahamtani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. .,Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
7
|
Huijbregts L, Aiello V, Soggia A, Ravassard P, Rachdi L, Scharfmann R, Albagli O. Culture, differentiation, and transduction of mouse E12.5 pancreatic spheres: an in vitro model for the secondary transition of pancreas development. Islets 2021; 13:10-23. [PMID: 33641620 PMCID: PMC8018339 DOI: 10.1080/19382014.2020.1863723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
During the secondary transition of rodent pancreatic development, mainly between E12.5 and E15.5 in mice, exocrine and endocrine populations differentiate from pancreatic progenitors. Here we describe an experimental system for its study in vitro. First, we show that spheres derived from dissociated E12.5 mouse pancreases differentiate within 7 days into most pancreatic exocrine and endocrine cell types, including beta cells. The proportion and spatial repartition of the different endocrine populations mirror those observed during normal development. Thus, dissociation and culture do not impair the developmental events affecting pancreatic progenitors during the secondary transition. Moreover, dissociated cells from mouse E12.5 pancreas were transduced with ecotropic MLV-based retroviral vectors or, though less efficiently, with a mixture of ALV(A)-based retroviral vectors and gesicles containing the TVA (Tumor Virus A) receptor. As an additional improvement, we also created a transgenic mouse line expressing TVA under the control of the 4.5 kB pdx1 promoter (pdx1-TVA). We demonstrate that pancreatic progenitors from dissociated pdx1-TVA pancreas can be specifically transduced by ALV(A)-based retroviral vectors. Using this model, we expressed an activated mutant of the YAP transcriptional co-activator in pancreatic progenitors. These experiments indicate that deregulated YAP activity reduces endocrine and exocrine differentiation in the resulting spheres, confirming and extending previously published data. Thus, our experimental model recapitulates in vitro the crucial developmental decisions arising at the secondary transition and provides a convenient tool to study their genetic control.
Collapse
Affiliation(s)
- Lukas Huijbregts
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris, France
| | - Virginie Aiello
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris, France
| | - Andrea Soggia
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris, France
| | - Philippe Ravassard
- Institut du Cerveau et de La Moelle Épinière (ICM), INSERM U1127, CNRS UMR 7225, Sorbonne Universités, Paris, France
| | - Latif Rachdi
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris, France
| | - Raphaël Scharfmann
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris, France
| | - Olivier Albagli
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris, France
- CONTACT Olivier Albagli Institut Cochin, INSERM U1016, 123 Bd Du Port-Royal, Paris75014, France
| |
Collapse
|
8
|
Tian JL, Gomeshtapeh FI. Potential Roles of O-GlcNAcylation in Primary Cilia- Mediated Energy Metabolism. Biomolecules 2020; 10:biom10111504. [PMID: 33139642 PMCID: PMC7693894 DOI: 10.3390/biom10111504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 10/30/2020] [Accepted: 10/30/2020] [Indexed: 12/26/2022] Open
Abstract
The primary cilium, an antenna-like structure on most eukaryotic cells, functions in transducing extracellular signals into intracellular responses via the receptors and ion channels distributed along it membrane. Dysfunction of this organelle causes an array of human diseases, known as ciliopathies, that often feature obesity and diabetes; this indicates the primary cilia's active role in energy metabolism, which it controls mainly through hypothalamic neurons, preadipocytes, and pancreatic β-cells. The nutrient sensor, O-GlcNAc, is widely involved in the regulation of energy homeostasis. Not only does O-GlcNAc regulate ciliary length, but it also modifies many components of cilia-mediated metabolic signaling pathways. Therefore, it is likely that O-GlcNAcylation (OGN) plays an important role in regulating energy homeostasis in primary cilia. Abnormal OGN, as seen in cases of obesity and diabetes, may play an important role in primary cilia dysfunction mediated by these pathologies.
Collapse
Affiliation(s)
- Jie L. Tian
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
- Correspondence: ; Tel.: +1-706-583-5551
| | | |
Collapse
|
9
|
Cozzitorto C, Mueller L, Ruzittu S, Mah N, Willnow D, Darrigrand JF, Wilson H, Khosravinia D, Mahmoud AA, Risolino M, Selleri L, Spagnoli FM. A Specialized Niche in the Pancreatic Microenvironment Promotes Endocrine Differentiation. Dev Cell 2020; 55:150-162.e6. [PMID: 32857951 PMCID: PMC7720791 DOI: 10.1016/j.devcel.2020.08.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/11/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022]
Abstract
The interplay between pancreatic epithelium and the surrounding microenvironment is pivotal for pancreas formation and differentiation as well as adult organ homeostasis. The mesenchyme is the main component of the embryonic pancreatic microenvironment, yet its cellular identity is broadly defined, and whether it comprises functionally distinct cell subsets is not known. Using genetic lineage tracing, transcriptome, and functional studies, we identified mesenchymal populations with different roles during pancreatic development. Moreover, we showed that Pbx transcription factors act within the mouse pancreatic mesenchyme to define a pro-endocrine specialized niche. Pbx directs differentiation of endocrine progenitors into insulin- and glucagon-positive cells through non-cell-autonomous regulation of ECM-integrin interactions and soluble molecules. Next, we measured functional conservation between mouse and human pancreatic mesenchyme by testing identified mesenchymal factors in an iPSC-based differentiation model. Our findings provide insights into how lineage-specific crosstalk between epithelium and neighboring mesenchymal cells underpin the generation of different pancreatic cell types.
Collapse
Affiliation(s)
- Corinna Cozzitorto
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Department of Ophthalmology & Department of Anatomy, Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laura Mueller
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Silvia Ruzittu
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Nancy Mah
- Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - David Willnow
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Jean-Francois Darrigrand
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Heather Wilson
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Daniel Khosravinia
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Amir-Ala Mahmoud
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Department of Orofacial Sciences & Department of Anatomy, University of California, San Francisco, 513 Parnassus Ave, HSW 710, San Francisco, CA 94143, USA
| | - Maurizio Risolino
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Department of Orofacial Sciences & Department of Anatomy, University of California, San Francisco, 513 Parnassus Ave, HSW 710, San Francisco, CA 94143, USA
| | - Licia Selleri
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Department of Orofacial Sciences & Department of Anatomy, University of California, San Francisco, 513 Parnassus Ave, HSW 710, San Francisco, CA 94143, USA
| | - Francesca M Spagnoli
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK.
| |
Collapse
|
10
|
Berthault C, Staels W, Scharfmann R. Purification of pancreatic endocrine subsets reveals increased iron metabolism in beta cells. Mol Metab 2020; 42:101060. [PMID: 32763423 PMCID: PMC7498953 DOI: 10.1016/j.molmet.2020.101060] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/23/2020] [Accepted: 07/30/2020] [Indexed: 11/18/2022] Open
Abstract
Objectives The main endocrine cell types in pancreatic islets are alpha, beta, and delta cells. Although these cell types have distinct roles in the regulation of glucose homeostasis, inadequate purification methods preclude the study of cell type-specific effects. We developed a reliable approach that enables simultaneous sorting of live alpha, beta, and delta cells from mouse islets for downstream analyses. Methods We developed an antibody panel against cell surface antigens to enable isolation of highly purified endocrine subsets from mouse islets based on the specific differential expression of CD71 on beta cells and CD24 on delta cells. We rigorously demonstrated the reliability and validity of our approach using bulk and single cell qPCR, immunocytochemistry, reporter mice, and transcriptomics. Results Pancreatic alpha, beta, and delta cells can be separated based on beta cell-specific CD71 surface expression and high expression of CD24 on delta cells. We applied our new sorting strategy to demonstrate that CD71, which is the transferrin receptor mediating the uptake of transferrin-bound iron, is upregulated in beta cells during early postnatal weeks. We found that beta cells express higher levels of several other genes implicated in iron metabolism and iron deprivation significantly impaired beta cell function. In human beta cells, CD71 is similarly required for iron uptake and CD71 surface expression is regulated in a glucose-dependent manner. Conclusions This study provides a novel and efficient purification method for murine alpha, beta, and delta cells, identifies for the first time CD71 as a postnatal beta cell-specific marker, and demonstrates a central role of iron metabolism in beta cell function. CD71 is a marker that is highly expressed in murine pancreatic beta-cells. CD71 and CD24 can be used to purify live murine alpha-, beta-, and delta-cells. Iron metabolism in murine beta-cells is increased compared to that in alpha-, and delta-cells. Human beta-cells regulate CD71 surface expression in a glucose-dependent manner.
Collapse
Affiliation(s)
- C Berthault
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 123 Boulevard de Port Royal, 75014 Paris, France.
| | - W Staels
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 123 Boulevard de Port Royal, 75014 Paris, France; Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, Belgium; Department of Pediatrics, Division of Pediatric Endocrinology, University Hospital of Brussels, Laarbeeklaan 101, Jette, Belgium
| | - R Scharfmann
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 123 Boulevard de Port Royal, 75014 Paris, France.
| |
Collapse
|
11
|
Villard O, Armanet M, Couderc G, Bony C, Moreaux J, Noël D, De Vos J, Klein B, Veyrune JL, Wojtusciszyn A. Characterization of immortalized human islet stromal cells reveals a MSC-like profile with pancreatic features. Stem Cell Res Ther 2020; 11:158. [PMID: 32303252 PMCID: PMC7165390 DOI: 10.1186/s13287-020-01649-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/12/2020] [Accepted: 03/10/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) represent an interesting tool to improve pancreatic islet transplantation. They have immunomodulatory properties and secrete supportive proteins. However, the functional properties of MSCs vary according to many factors such as donor characteristics, tissue origin, or isolation methods. To counteract this heterogeneity, we aimed to immortalize and characterize adherent cells derived from human pancreatic islets (hISCs), using phenotypic, transcriptomic, and functional analysis. METHODS Adherent cells derived from human islets in culture were infected with a hTERT retrovirus vector and then characterized by microarray hybridization, flow cytometry analysis, and immunofluorescence assays. Osteogenic, adipogenic, and chondrogenic differentiation as well as PBMC proliferation suppression assays were used to compare the functional abilities of hISCs and MSCs. Extracellular matrix (ECM) gene expression profile analysis was performed using the SAM (Significance Analysis of Microarrays) software, and protein expression was confirmed by western blotting. RESULTS hISCs kept an unlimited proliferative potential. They exhibited several properties of MSCs such as CD73, CD90, and CD105 expression and differentiation capacity. From a functional point of view, hISCs inhibited the proliferation of activated peripheral blood mononuclear cells. The transcriptomic profile of hISCs highly clusterized with bone marrow (BM)-MSCs and revealed a differential enrichment of genes involved in the organization of the ECM. Indeed, the expression and secretion profiles of ECM proteins including collagens I, IV, and VI, fibronectin, and laminins, known to be expressed in abundance around and within the islets, were different between hISCs and BM-MSCs. CONCLUSION We generated a new human cell line from pancreatic islets, with MSCs properties and retaining some pancreatic specificities related to the production of ECM proteins. hISCs appear as a very promising tool in islet transplantation by their availability (as a source of inexhaustible source of cells) and ability to secrete a supportive "pancreatic" microenvironment.
Collapse
Affiliation(s)
- Orianne Villard
- Laboratory of Cell Therapy for Diabetes, Institute of Regenerative Medicine and Biotherapy, Univ. Montpellier, CHU Montpellier, Montpellier, France
- Department of Endocrinology, Diabetes, and Nutrition, Univ. Montpellier, CHU Montpellier, Montpellier, France
| | - Mathieu Armanet
- Laboratory of Cell Therapy for Diabetes, Institute of Regenerative Medicine and Biotherapy, Univ. Montpellier, CHU Montpellier, Montpellier, France
- Cell Therapy Unit, Hospital Saint- Louis, AP-HP, Paris, France
- Department of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, 8 avenue de la Sallaz - 1011, Lausanne, Switzerland
| | - Guilhem Couderc
- Department of Biological Haematology, Univ. Montpellier, CHU Montpellier, Montpellier, France
- Department of Cell and Tissue Engineering, Univ. Montpellier, CHU Montpellier, Montpellier, France
| | - Claire Bony
- IRMB, INSERM U 1183, Univ Montpellier, INSERM, Montpellier, France
| | - Jerome Moreaux
- Department of Biological Haematology, Univ. Montpellier, CHU Montpellier, Montpellier, France
- IGH, Univ Montpellier, CNRS, Montpellier, France
| | - Daniele Noël
- IRMB, INSERM U 1183, Univ Montpellier, INSERM, Montpellier, France
| | - John De Vos
- Department of Biological Haematology, Univ. Montpellier, CHU Montpellier, Montpellier, France
- Department of Cell and Tissue Engineering, Univ. Montpellier, CHU Montpellier, Montpellier, France
- IRMB, INSERM U 1183, Univ Montpellier, INSERM, Montpellier, France
| | - Bernard Klein
- Department of Cell and Tissue Engineering, Univ. Montpellier, CHU Montpellier, Montpellier, France
| | - Jean-Luc Veyrune
- Department of Biological Haematology, Univ. Montpellier, CHU Montpellier, Montpellier, France
- Department of Cell and Tissue Engineering, Univ. Montpellier, CHU Montpellier, Montpellier, France
| | - Anne Wojtusciszyn
- Laboratory of Cell Therapy for Diabetes, Institute of Regenerative Medicine and Biotherapy, Univ. Montpellier, CHU Montpellier, Montpellier, France.
- Department of Endocrinology, Diabetes, and Nutrition, Univ. Montpellier, CHU Montpellier, Montpellier, France.
- Department of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, 8 avenue de la Sallaz - 1011, Lausanne, Switzerland.
| |
Collapse
|
12
|
Sufu- and Spop-mediated downregulation of Hedgehog signaling promotes beta cell differentiation through organ-specific niche signals. Nat Commun 2019; 10:4647. [PMID: 31604927 PMCID: PMC6789033 DOI: 10.1038/s41467-019-12624-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 09/20/2019] [Indexed: 12/22/2022] Open
Abstract
Human embryonic stem cell-derived beta cells offer a promising cell-based therapy for diabetes. However, efficient stem cell to beta cell differentiation has proven difficult, possibly due to the lack of cross-talk with the appropriate mesenchymal niche. To define organ-specific niche signals, we isolated pancreatic and gastrointestinal stromal cells, and analyzed their gene expression during development. Our genetic studies reveal the importance of tightly regulated Hedgehog signaling in the pancreatic mesenchyme: inactivation of mesenchymal signaling leads to annular pancreas, whereas stroma-specific activation of signaling via loss of Hedgehog regulators, Sufu and Spop, impairs pancreatic growth and beta cell genesis. Genetic rescue and transcriptome analyses show that these Sufu and Spop knockout defects occur through Gli2-mediated activation of gastrointestinal stromal signals such as Wnt ligands. Importantly, inhibition of Wnt signaling in organoid and human stem cell cultures significantly promotes insulin-producing cell generation, altogether revealing the requirement for organ-specific regulation of stromal niche signals.
Collapse
|
13
|
Sakhneny L, Khalifa-Malka L, Landsman L. Pancreas organogenesis: Approaches to elucidate the role of epithelial-mesenchymal interactions. Semin Cell Dev Biol 2019; 92:89-96. [DOI: 10.1016/j.semcdb.2018.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 08/26/2018] [Accepted: 08/27/2018] [Indexed: 12/12/2022]
|
14
|
Soltanian A, Ghezelayagh Z, Mazidi Z, Halvaei M, Mardpour S, Ashtiani MK, Hajizadeh-Saffar E, Tahamtani Y, Baharvand H. Generation of functional human pancreatic organoids by transplants of embryonic stem cell derivatives in a 3D-printed tissue trapper. J Cell Physiol 2019; 234:9564-9576. [PMID: 30362564 DOI: 10.1002/jcp.27644] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/02/2018] [Indexed: 12/12/2022]
Abstract
Organoids can be regarded as a beneficial tool for discovery of new therapeutics for diabetes and/or maturation of pancreatic progenitors (PP) towards β cells. Here, we devised a strategy to enhance maturation of PP by assembly of three-dimensional (3D) pancreatic organoids (PO) containing human embryonic stem (ES) cell derivatives including ES-derived pancreatic duodenal homeobox 1 (PDX1) + early PP, mesenchymal stem cells, and endothelial cells at an optimized cell ratio, on Matrigel. The PO was placed in a 3D-printed tissue trapper and heterotopically implanted into the peritoneal cavity of immunodeficient mice where it remained for 90 days. Our results indicated that, in contrast to corresponding early PP transplants, 3D PO developed more vascularization as indicated by greater area and number of vessels, a higher number of insulin-positive cells and improvement of human C-peptide secretions. Based on our findings, PO-derived β cells could be considered a novel strategy to study human β-cell development, novel therapeutics, and regenerative medicine for diabetes.
Collapse
Affiliation(s)
- Anahita Soltanian
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Ghezelayagh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Zahra Mazidi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Majid Halvaei
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Soura Mardpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Kazemi Ashtiani
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Yaser Tahamtani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
15
|
Huijbregts L, Petersen MBK, Berthault C, Hansson M, Aiello V, Rachdi L, Grapin-Botton A, Honore C, Scharfmann R. Bromodomain and Extra Terminal Protein Inhibitors Promote Pancreatic Endocrine Cell Fate. Diabetes 2019; 68:761-773. [PMID: 30655386 DOI: 10.2337/db18-0224] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 01/07/2019] [Indexed: 11/13/2022]
Abstract
Bromodomain and extraterminal (BET) proteins are epigenetic readers that interact with acetylated lysines of histone tails. Recent studies have demonstrated their role in cancer progression because they recruit key components of the transcriptional machinery to modulate gene expression. However, their role during embryonic development of the pancreas has never been studied. Using mouse embryonic pancreatic explants and human induced pluripotent stem cells (hiPSCs), we show that BET protein inhibition with I-BET151 or JQ1 enhances the number of neurogenin3 (NEUROG3) endocrine progenitors. In mouse explants, BET protein inhibition further led to increased expression of β-cell markers but in the meantime, strongly downregulated Ins1 expression. Similarly, although acinar markers, such as Cpa1 and CelA, were upregulated, Amy expression was repressed. In hiPSCs, BET inhibitors strongly repressed C-peptide and glucagon during endocrine differentiation. Explants and hiPSCs were then pulsed with BET inhibitors to increase NEUROG3 expression and further chased without inhibitors. Endocrine development was enhanced in explants with higher expression of insulin and maturation markers, such as UCN3 and MAFA. In hiPSCs, the outcome was different because C-peptide expression remained lower than in controls, but ghrelin expression was increased. Altogether, by using two independent models of pancreatic development, we show that BET proteins regulate multiple aspects of pancreatic development.
Collapse
Affiliation(s)
- Lukas Huijbregts
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
| | - Maja Borup Kjær Petersen
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Claire Berthault
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
| | | | - Virginie Aiello
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
| | - Latif Rachdi
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
| | - Anne Grapin-Botton
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Christian Honore
- Department of Stem Cell Biology, Novo Nordisk A/S, Måløv, Denmark
| | - Raphael Scharfmann
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
| |
Collapse
|
16
|
Pancreas organogenesis: The interplay between surrounding microenvironment(s) and epithelium-intrinsic factors. Curr Top Dev Biol 2019; 132:221-256. [DOI: 10.1016/bs.ctdb.2018.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
17
|
Abstract
Stellate cells are resident lipid-storing cells of the pancreas and liver that transdifferentiate to a myofibroblastic state in the context of tissue injury. Beyond having roles in tissue homeostasis, stellate cells are increasingly implicated in pathological fibrogenic and inflammatory programs that contribute to tissue fibrosis and that constitute a growth-permissive tumor microenvironment. Although the capacity of stellate cells for extracellular matrix production and remodeling has long been appreciated, recent research efforts have demonstrated diverse roles for stellate cells in regulation of epithelial cell fate, immune modulation, and tissue health. Our present understanding of stellate cell biology in health and disease is discussed here, as are emerging means to target these multifaceted cells for therapeutic benefit.
Collapse
Affiliation(s)
- Mara H Sherman
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon 97201, USA;
| |
Collapse
|
18
|
Jimenez-Caliani AJ, Pillich R, Yang W, Diaferia GR, Meda P, Crisa L, Cirulli V. αE-Catenin Is a Positive Regulator of Pancreatic Islet Cell Lineage Differentiation. Cell Rep 2018; 20:1295-1306. [PMID: 28793255 PMCID: PMC5611824 DOI: 10.1016/j.celrep.2017.07.035] [Citation(s) in RCA: 14] [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: 12/28/2016] [Revised: 05/09/2017] [Accepted: 07/13/2017] [Indexed: 01/13/2023] Open
Abstract
The development and function of epithelia depend on the establishment and maintenance of cell-cell adhesion and intercellular junctions, which operate as mechanosensor hubs for the transduction of biochemical signals regulating cell proliferation, differentiation, survival, and regeneration. Here, we show that αE-catenin, a key component of adherens junctions, functions as a positive regulator of pancreatic islet cell lineage differentiation by repressing the sonic hedgehog pathway (SHH). Thus, deletion of αE-catenin in multipotent pancreatic progenitors resulted in (1) loss of adherens junctions, (2) constitutive activation of SHH, (3) decrease in islet cell lineage differentiation, and (4) accumulation of immature Sox9+ progenitors. Pharmacological blockade of SHH signaling in pancreatic organ cultures and in vivo rescued this defect, allowing αE-catenin-null Sox9+ pancreatic progenitors to differentiate into endocrine cells. The results uncover crucial functions of αE-catenin in pancreatic islet development and harbor significant implications for the design of β cell replacement and regeneration therapies in diabetes.
Collapse
Affiliation(s)
- Antonio J Jimenez-Caliani
- Department of Medicine, UW Diabetes Institute, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Dermatology, Rheumatology, Diabetology, University of Bremen, Bremen, Germany
| | - Rudolf Pillich
- Department of Medicine, UW Diabetes Institute, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Wendy Yang
- Department of Medicine, UW Diabetes Institute, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Giuseppe R Diaferia
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Laura Crisa
- Department of Medicine, UW Diabetes Institute, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| | - Vincenzo Cirulli
- Department of Medicine, UW Diabetes Institute, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
19
|
Angelo JR, Tremblay KD. Identification and fate mapping of the pancreatic mesenchyme. Dev Biol 2018; 435:15-25. [PMID: 29329912 DOI: 10.1016/j.ydbio.2018.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 01/06/2018] [Accepted: 01/06/2018] [Indexed: 12/25/2022]
Abstract
The murine pancreas buds from the ventral embryonic endoderm at approximately 8.75 dpc and a second pancreas bud emerges from the dorsal endoderm by 9.0 dpc. Although it is clear that secreted signals from adjacent mesoderm-derived sources are required for both the appropriate emergence and further refinement of the pancreatic endoderm, neither the exact signals nor the requisite tissue sources have been defined in mammalian systems. Herein we use DiI fate mapping of cultured murine embryos to identify the embryonic sources of both the early inductive and later condensed pancreatic mesenchyme. Despite being capable of supporting pancreas induction from dorsal endoderm in co-culture experiments, we find that in the context of the developing embryo, the dorsal aortae as well as the paraxial, intermediate, and lateral mesoderm derivatives only transiently associate with the dorsal pancreas bud, producing descendants that are decidedly anterior to the pancreas bud. Unlike these other mesoderm derivatives, the axial (notochord) descendants maintain association with the dorsal pre-pancreatic endoderm and early pancreas bud. This fate mapping data points to the notochord as the likely inductive source in vivo while also revealing dynamic morphogenetic movements displayed by individual mesodermal subtypes. Because none of the mesoderm examined above produced the pancreatic mesenchyme that condenses around the induced bud to support exocrine and endocrine differentiation, we also sought to identify the mesodermal origins of this mesenchyme. We identify a portion of the coelomic mesoderm that contributes to the condensed pancreatic mesenchyme. In conclusion, we identify a portion of the notochord as a likely source of the signals required to induce and maintain the early dorsal pancreas bud, demonstrate that the coelomic mesothelium contributes to the dorsal and ventral pancreatic mesenchyme, and provide insight into the dynamic morphological rearrangements of mesoderm-derived tissues during early organogenesis stages of mammalian development.
Collapse
Affiliation(s)
- Jesse R Angelo
- Department of Veterinary&Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Kimberly D Tremblay
- Department of Veterinary&Animal Sciences, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
20
|
Broche B, Ben Fradj S, Aguilar E, Sancerni T, Bénard M, Makaci F, Berthault C, Scharfmann R, Alves-Guerra MC, Duvillié B. Mitochondrial Protein UCP2 Controls Pancreas Development. Diabetes 2018; 67:78-84. [PMID: 29079704 DOI: 10.2337/db17-0118] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 10/23/2017] [Indexed: 11/13/2022]
Abstract
The mitochondrial carrier uncoupling protein (UCP) 2 belongs to the family of the UCPs. Despite its name, it is now accepted that UCP2 is rather a metabolite transporter than a UCP. UCP2 can regulate oxidative stress and/or energetic metabolism. In rodents, UCP2 is involved in the control of α- and β-cell mass as well as insulin and glucagon secretion. Our aim was to determine whether the effects of UCP2 observed on β-cell mass have an embryonic origin. Thus, we used Ucp2 knockout mice. We found an increased size of the pancreas in Ucp2-/- fetuses at embryonic day 16.5, associated with a higher number of α- and β-cells. This phenotype was caused by an increase of PDX1+ progenitor cells. Perinatally, an increase in the proliferation of endocrine cells also participates in their expansion. Next, we analyzed the oxidative stress in the pancreata. We quantified an increased nuclear translocation of nuclear factor erythroid 2-related factor 2 (NRF2) in the mutant, suggesting an increased production of reactive oxygen species (ROS). Phosphorylation of AKT, an ROS target, was also activated in the Ucp2-/- pancreata. Finally, administration of the antioxidant N-acetyl-l-cysteine to Ucp2-/- pregnant mice alleviated the effect of knocking out UCP2 on pancreas development. Together, these data demonstrate that UCP2 controls pancreas development through the ROS-AKT signaling pathway.
Collapse
Affiliation(s)
- Benjamin Broche
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Selma Ben Fradj
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Esther Aguilar
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Tiphaine Sancerni
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Matthieu Bénard
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Fatna Makaci
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Claire Berthault
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Raphaël Scharfmann
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marie-Clotilde Alves-Guerra
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bertrand Duvillié
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
21
|
Ameri J, Borup R, Prawiro C, Ramond C, Schachter KA, Scharfmann R, Semb H. Efficient Generation of Glucose-Responsive Beta Cells from Isolated GP2 + Human Pancreatic Progenitors. Cell Rep 2017; 19:36-49. [PMID: 28380361 DOI: 10.1016/j.celrep.2017.03.032] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 02/10/2017] [Accepted: 03/09/2017] [Indexed: 12/29/2022] Open
Abstract
Stem cell-based therapy for type 1 diabetes would benefit from implementation of a cell purification step at the pancreatic endoderm stage. This would increase the safety of the final cell product, allow the establishment of an intermediate-stage stem cell bank, and provide a means for upscaling β cell manufacturing. Comparative gene expression analysis revealed glycoprotein 2 (GP2) as a specific cell surface marker for isolating pancreatic endoderm cells (PECs) from differentiated hESCs and human fetal pancreas. Isolated GP2+ PECs efficiently differentiated into glucose responsive insulin-producing cells in vitro. We found that in vitro PEC proliferation declines due to enhanced expression of the cyclin-dependent kinase (CDK) inhibitors CDKN1A and CDKN2A. However, we identified a time window when reducing CDKN1A or CDKN2A expression increased proliferation and yield of GP2+ PECs. Altogether, our results contribute tools and concepts toward the isolation and use of PECs as a source for the safe production of hPSC-derived β cells.
Collapse
Affiliation(s)
- Jacqueline Ameri
- The Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark; Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, BMC, B10, 22184 Lund, Sweden
| | - Rehannah Borup
- Center for Genomic Medicine, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Christy Prawiro
- The Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Cyrille Ramond
- INSERM U1016, University Paris-Descartes, Cochin Institute, 75014 Paris, France
| | - Karen A Schachter
- The Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Raphael Scharfmann
- INSERM U1016, University Paris-Descartes, Cochin Institute, 75014 Paris, France
| | - Henrik Semb
- The Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark; Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, BMC, B10, 22184 Lund, Sweden.
| |
Collapse
|
22
|
Bastidas-Ponce A, Scheibner K, Lickert H, Bakhti M. Cellular and molecular mechanisms coordinating pancreas development. Development 2017; 144:2873-2888. [PMID: 28811309 DOI: 10.1242/dev.140756] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The pancreas is an endoderm-derived glandular organ that participates in the regulation of systemic glucose metabolism and food digestion through the function of its endocrine and exocrine compartments, respectively. While intensive research has explored the signaling pathways and transcriptional programs that govern pancreas development, much remains to be discovered regarding the cellular processes that orchestrate pancreas morphogenesis. Here, we discuss the developmental mechanisms and principles that are known to underlie pancreas development, from induction and lineage formation to morphogenesis and organogenesis. Elucidating such principles will help to identify novel candidate disease genes and unravel the pathogenesis of pancreas-related diseases, such as diabetes, pancreatitis and cancer.
Collapse
Affiliation(s)
- Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.,Technical University of Munich, Medical Faculty, 81675 Munich, Germany
| | - Katharina Scheibner
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.,Technical University of Munich, Medical Faculty, 81675 Munich, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.,Technical University of Munich, Medical Faculty, 81675 Munich, Germany
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany .,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
| |
Collapse
|
23
|
Scavuzzo MA, Yang D, Borowiak M. Organotypic pancreatoids with native mesenchyme develop Insulin producing endocrine cells. Sci Rep 2017; 7:10810. [PMID: 28883507 PMCID: PMC5589819 DOI: 10.1038/s41598-017-11169-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 08/15/2017] [Indexed: 12/19/2022] Open
Abstract
Replacement of lost beta cells in patients with diabetes has the potential to alleviate them of their disease, yet current protocols to make beta cells are inadequate for therapy. In vitro screens can reveal the signals necessary for endocrine maturation to improve beta cell production, however the complexities of in vivo development that lead to beta cell formation are lost in two-dimensional systems. Here, we create three-dimensional organotypic pancreatic cultures, named pancreatoids, composed of embryonic day 10.5 murine epithelial progenitors and native mesenchyme. These progenitors assemble in scaffold-free, floating conditions and, with the inclusion of native mesenchyme, develop into pancreatoids expressing markers of different pancreatic lineages including endocrine-like cells. Treatment of pancreatoids with (-)-Indolactam-V or phorbol 12-myristate 13-acetate, two protein kinase C activators, leads to altered morphology which otherwise would be overlooked in two-dimensional systems. Protein kinase C activation also led to fewer Insulin+ cells, decreased Ins1 and Ins2 mRNA levels, and increased Pdx1 and Hes1 mRNA levels with a high number of DBA+ cells. Thus, organotypic pancreatoids provide a useful tool for developmental studies, and can further be used for disease modeling, small molecules and genetic screens, or applied to human pluripotent stem cell differentiation for beta-like cell formation.
Collapse
Affiliation(s)
- Marissa A Scavuzzo
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Diane Yang
- Molecular and Cellular Biology Department, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Malgorzata Borowiak
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Molecular and Cellular Biology Department, Baylor College of Medicine, Houston, TX, 77030, USA. .,Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX, 77030, USA. .,Stem Cell and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, 77030, USA. .,McNair Medical Institute, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
24
|
Epshtein A, Rachi E, Sakhneny L, Mizrachi S, Baer D, Landsman L. Neonatal pancreatic pericytes support β-cell proliferation. Mol Metab 2017; 6:1330-1338. [PMID: 29031732 PMCID: PMC5641631 DOI: 10.1016/j.molmet.2017.07.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE The maintenance and expansion of β-cell mass rely on their proliferation, which reaches its peak in the neonatal stage. β-cell proliferation was found to rely on cells of the islet microenvironment. We hypothesized that pericytes, which are components of the islet vasculature, support neonatal β-cell proliferation. METHODS To test our hypothesis, we combined in vivo and in vitro approaches. Briefly, we used a Diphtheria toxin-based transgenic mouse system to specifically deplete neonatal pancreatic pericytes in vivo. We further cultured neonatal pericytes isolated from the neonatal pancreas and combined the use of a β-cell line and primary cultured mouse β-cells. RESULTS Our findings indicate that neonatal pancreatic pericytes are required and sufficient for β-cell proliferation. We observed impaired proliferation of neonatal β-cells upon in vivo depletion of pancreatic pericytes. Furthermore, exposure to pericyte-conditioned medium stimulated proliferation in cultured β-cells. CONCLUSIONS This study introduces pancreatic pericytes as regulators of neonatal β-cell proliferation. In addition to advancing current understanding of the physiological β-cell replication process, these findings could facilitate the development of protocols aimed at expending these cells as a potential cure for diabetes.
Collapse
Affiliation(s)
- Alona Epshtein
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eleonor Rachi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lina Sakhneny
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shani Mizrachi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daria Baer
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Limor Landsman
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
25
|
Kaitsuka T, Kobayashi K, Otsuka W, Kubo T, Hakim F, Wei FY, Shiraki N, Kume S, Tomizawa K. Erythropoietin facilitates definitive endodermal differentiation of mouse embryonic stem cells via activation of ERK signaling. Am J Physiol Cell Physiol 2017; 312:C573-C582. [PMID: 28298334 DOI: 10.1152/ajpcell.00071.2016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 03/06/2017] [Accepted: 03/06/2017] [Indexed: 01/07/2023]
Abstract
Artificially generated pancreatic β-cells from pluripotent stem cells are expected for cell replacement therapy for type 1 diabetes. Several strategies are adopted to direct pluripotent stem cells toward pancreatic differentiation. However, a standard differentiation method for clinical application has not been established. It is important to develop more effective and safer methods for generating pancreatic β-cells without toxic or mutagenic chemicals. In the present study, we screened several endogenous factors involved in organ development to identify the factor, which induced the efficiency of pancreatic differentiation and found that treatment with erythropoietin (EPO) facilitated the differentiation of mouse embryonic stem cells (ESCs) into definitive endoderm. At an early stage of differentiation, EPO treatment significantly increased Sox17 gene expression, as a marker of the definitive endoderm. Contrary to the canonical function of EPO, it did not affect the levels of phosphorylated JAK2 and STAT5, but stimulated the phosphorylation of ERK1/2 and Akt. The MEK inhibitor U0126 significantly inhibited EPO-induced Sox17 expression. The differentiation of ESCs into definitive endoderm is an important step for the differentiation into pancreatic and other endodermal lineages. This study suggests a possible role of EPO in embryonic endodermal development and a new agent for directing the differentiation into endodermal lineages like pancreatic β-cells.
Collapse
Affiliation(s)
- Taku Kaitsuka
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kohei Kobayashi
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Wakako Otsuka
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takuya Kubo
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Farzana Hakim
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Fan-Yan Wei
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Nobuaki Shiraki
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan; and.,Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| | - Shoen Kume
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan; and.,Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| | - Kazuhito Tomizawa
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan;
| |
Collapse
|
26
|
Samadi R, Shafiei B, Azizi F, Ghasemi A. Radioactive Iodine Therapy and Glucose Tolerance. CELL JOURNAL 2017; 19:184-193. [PMID: 28670511 PMCID: PMC5413587 DOI: 10.22074/cellj.2016.4251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/19/2016] [Indexed: 11/09/2022]
Abstract
Radioactive iodine therapy is commonly used as an adjuvant therapy in follicular and
papillary thyroid carcinoma (PTC) and in the treatment of Graves’ disease (GD). The
basis of this therapy is the accumulation of radioactive iodine by the sodium-iodide
symporter (NIS) in the thyroid gland. Expression of NIS by extrathyroidal tissues such
as islets of pancreas has been reported. Radioactive iodine uptake by pancreatic
beta-cells can potentially damage these cells. In this study, we discuss the possible
associations between radioactive iodine and glucose intolerance. Overall, radioactive
iodine uptake by the pancreas may damage beta-cells and predispose patients to
glucose intolerance or type 2 diabetes, particularly in patients exposed to radioactive
iodine therapy following total thyroidectomy. Further studies are needed to clarify and
confirm this association.
Collapse
Affiliation(s)
- Roghaieh Samadi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Shafiei
- Department of Nuclear Medicine, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Al Madhoun A, Ali H, AlKandari S, Atizado VL, Akhter N, Al-Mulla F, Atari M. Defined three-dimensional culture conditions mediate efficient induction of definitive endoderm lineage from human umbilical cord Wharton's jelly mesenchymal stem cells. Stem Cell Res Ther 2016; 7:165. [PMID: 27852316 PMCID: PMC5111269 DOI: 10.1186/s13287-016-0426-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 10/18/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) are gaining increasing interest as an alternative source of stem cells for regenerative medicine applications. Definitive endoderm (DE) specification is a prerequisite for the development of vital organs such as liver and pancreas. Hence, efficient induction of the DE lineage from stem cells is crucial for subsequent generation of clinically relevant cell types. Here we present a defined 3D differentiation protocol of WJ-MSCs into DE cells. METHODS WJ-MSCs were cultured in suspension to generate spheroids, about 1500 cells each, for 7 days. The serum-free differentiation media contained specific growth factors, cytokines, and small molecules that specifically regulate signaling pathways including sonic hedgehog, bone morphogenetic protein, Activin/Wnt, and Notch. RESULTS We obtained more than 85 % DE cells as shown with FACS analysis using antibodies directed against the DE marker CXCR4. In addition, biochemical and molecular analysis of bona-fide DE markers revealed a time-course induction of Sox17, CXCR4, and FoxA2. Focused PCR-based array also indicated a specific induction into the DE lineage. CONCLUSIONS In this study, we report an efficient serum-free protocol to differentiate WJ-MSCs into DE cells utilizing 3D spheroid formation. Our approach might aid in the development of new protocols to obtain DE-derivative lineages including liver-like and pancreatic insulin-producing cells.
Collapse
Affiliation(s)
| | - Hamad Ali
- Research Division, Dasman Diabetes Institute, 1180 Dasman, Kuwait
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center, Kuwait University, Al-Jabriya, Kuwait
| | - Sarah AlKandari
- Research Division, Dasman Diabetes Institute, 1180 Dasman, Kuwait
| | | | - Nadeem Akhter
- Research Division, Dasman Diabetes Institute, 1180 Dasman, Kuwait
| | - Fahd Al-Mulla
- Department of Pathology, Molecular Pathology Unit, Faculty of Medicine, Health Sciences Center, Kuwait University, Al-Jabriya, Kuwait
| | - Maher Atari
- UIC Regenerative Medicine Research Institute, International University of Catalonia, Barcelona, Spain
| |
Collapse
|
28
|
Hox6 genes modulate in vitro differentiation of mESCs to insulin-producing cells. In Vitro Cell Dev Biol Anim 2016; 52:974-982. [PMID: 27444630 DOI: 10.1007/s11626-016-0066-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 06/08/2016] [Indexed: 01/19/2023]
Abstract
The differentiation of glucose-responsive, insulin-producing cells from ESCs in vitro is promising as a cellular therapy for the treatment of diabetes, a devastating and common disease. Pancreatic β-cells are derived from the endoderm in vivo and therefore most current protocols attempt to generate a pure population of first endoderm, then pancreas epithelium, and finally insulin-producing cells. Despite this, differentiation protocols result in mixed populations of cells that are often poorly defined, but also contain mesoderm. Using an in vitro mESC-to-β cell differentiation protocol, we show that expression of region-specific Hox genes is induced. We also show that the loss of function of the Hox6 paralogous group, genes expressed only in the mesenchyme of the pancreas (not epithelium), affect the differentiation of insulin-producing cells in vitro. This work is consistent with the important role for these mesoderm-specific factors in vivo and highlights contribution of supporting mesenchymal cells in in vitro differentiation.
Collapse
|
29
|
Lecomte MJ, Pechberty S, Machado C, Da Barroca S, Ravassard P, Scharfmann R, Czernichow P, Duvillié B. Aggregation of Engineered Human β-Cells Into Pseudoislets: Insulin Secretion and Gene Expression Profile in Normoxic and Hypoxic Milieu. CELL MEDICINE 2016; 8:99-112. [PMID: 28003935 DOI: 10.3727/215517916x692843] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Innovative treatments to cure type 1 diabetes are being actively researched. Among the different strategies, the replacement of β-cells has given promising results. Classically, islets from cadaveric donors are transplanted into diabetic patients, but recently phase I clinical trials that use stem cell-derived β-cells have been started. Such protocols require either an immunosuppressive treatment or the macroencapsulation of the β-cells. They involve cell aggregation and the exposure of the cells to hypoxia. Using an engineered human β-cell, we have addressed these two problems: a novel human β-cell line called EndoC-βH3 was cultured as single cells or aggregated clusters. EndoC-βH3 cells were also cultured at normal atmospheric oxygen tension (pO2 = 21%) or hypoxia (pO2 = 3%) in the presence or absence of modulators of the hypoxia-inducible factor 1α (HIF1α) pathway. Cell aggregation improved glucose-stimulated insulin secretion, demonstrating the benefit of cell-cell contacts. Low oxygen tension decreased β-cell viability and their sensitivity to glucose, but did not alter insulin production nor the insulin secretion capacity of the remaining cells. To investigate the role of HIF1α, we first used a HIF stabilizer at pO2 = 21%. This led to a mild decrease in cell viability, impaired glucose sensitivity, and altered insulin secretion. Finally, we used a HIF inhibitor on EndoC-βH3 pseudoislets exposed to hypoxia. Such treatment considerably decreased cell viability. In conclusion, aggregation of the EndoC-βH3 cells seems to be important to improve their function. A fraction of the EndoC-βH3 cells are resistant to hypoxia, depending on the level of activity of HIF1α. Thus, these cells represent a good human cell model for future investigations on islet cell transplantation analysis.
Collapse
Affiliation(s)
- Marie-José Lecomte
- Univercell-Biosolutions, Centre de recherche des Cordeliers , Paris , France
| | - Séverine Pechberty
- Univercell-Biosolutions, Centre de recherche des Cordeliers , Paris , France
| | - Cécile Machado
- Univercell-Biosolutions, Centre de recherche des Cordeliers , Paris , France
| | - Sandra Da Barroca
- Univercell-Biosolutions, Centre de recherche des Cordeliers , Paris , France
| | - Philippe Ravassard
- † Sorbonne Universités, UPMC Univ Paris 06, Inserm, CNRS, Institut du cerveau et de la moelle (ICM)-Hôpital Pitié-Salpêtrière , Paris , France
| | - Raphaël Scharfmann
- ‡INSERM U1016, Institut Cochin, Paris, France; §Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Paul Czernichow
- Univercell-Biosolutions, Centre de recherche des Cordeliers , Paris , France
| | - Bertrand Duvillié
- ‡INSERM U1016, Institut Cochin, Paris, France; §Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| |
Collapse
|
30
|
Proinflammatory Cytokines Induce Endocrine Differentiation in Pancreatic Ductal Cells via STAT3-Dependent NGN3 Activation. Cell Rep 2016; 15:460-470. [PMID: 27068459 DOI: 10.1016/j.celrep.2016.03.036] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/18/2016] [Accepted: 03/09/2016] [Indexed: 12/16/2022] Open
Abstract
A major goal of diabetes research is to develop strategies that replenish pancreatic insulin-producing beta cells. One emerging strategy is to harness pancreatic plasticity-the ability of pancreatic cells to undergo cellular interconversions-a phenomenon implicated in physiological stress and pancreatic injury. Here, we investigate the effects of inflammatory cytokine stress on the differentiation potential of ductal cells in a human cell line, in mouse ductal cells by pancreatic intraductal injection, and during the progression of autoimmune diabetes in the non-obese diabetic (NOD) mouse model. We find that inflammatory cytokine insults stimulate epithelial-to-mesenchymal transition (EMT) as well as the endocrine program in human pancreatic ductal cells via STAT3-dependent NGN3 activation. Furthermore, we show that inflammatory cytokines activate ductal-to-endocrine cell reprogramming in vivo independent of hyperglycemic stress. Together, our findings provide evidence that inflammatory cytokines direct ductal-to-endocrine cell differentiation, with implications for beta cell regeneration.
Collapse
|
31
|
Liang J, Wu SY, Zhang D, Wang L, Leung KK, Leung PS. NADPH Oxidase-Dependent Reactive Oxygen Species Stimulate β-Cell Regeneration Through Differentiation of Endocrine Progenitors in Murine Pancreas. Antioxid Redox Signal 2016; 24:419-33. [PMID: 26464216 DOI: 10.1089/ars.2014.6135] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIMS Reactive oxygen species (ROS) act as second messengers for redox modification of transcription factors essential for differentiation. The nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, a major source of ROS, has been shown to regulate differentiation of various progenitor cells, while its role in pancreatic endocrine cell differentiation is unclear. This study was aimed at this knowledge gap. RESULTS Our results showed that ROS levels were dynamically changed during pancreas development concomitant with endocrine cell differentiation induced by modest exogenous ROS in rudiment cultures. NOX4, but not NOX2, the member of NADPH oxidase, was expressed persistently in endocrine lineage and showed high activity in critical pancreas development phase. Inhibition of NADPH oxidase activity impeded the differentiation of endocrine progenitors in vitro, and exogenous ROS reversed this effect. Studies performed in streptozotocin (STZ)-injected neonatal rats showed that diphenyleneiodonium (DPI) obstructed β-cell regeneration through the suppression of neurogenin 3 (NGN3) expression, but not Ki67-labeling β-cells, indicating that ROS stimulation promoted differentiation beyond proliferation of β-cells. Inhibition of NADPH oxidase also reduced expression of SRY (sex-determining region Y)-box 9 (SOX9), a transcriptional regulator of Ngn3, in endocrine precursor cells, both in vivo and in vitro. Overexpression of SOX9 attenuated the reduction of NGN3 induced by suppression of NADPH oxidase. INNOVATION AND CONCLUSION This is the first study to demonstrate NADPH oxidase, especially NOX4-dependent ROS that promotes pancreatic progenitor cell differentiation into endocrine cells both in vitro and in vivo, probably through the regulation of SOX9. We provide evidence that NADPH oxidase-dependent ROS-mediated signaling is necessary for endocrine cell differentiation, which provides a potential strategy for efficient generation of insulin-producing cells in clinical application.
Collapse
Affiliation(s)
- Juan Liang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong , Hong Kong, China
| | - Shang Ying Wu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong , Hong Kong, China
| | - Dan Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong , Hong Kong, China
| | - Lin Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong , Hong Kong, China
| | - Kwan Keung Leung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong , Hong Kong, China
| | - Po Sing Leung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong , Hong Kong, China
| |
Collapse
|
32
|
Kofent J, Spagnoli FM. Xenopus as a model system for studying pancreatic development and diabetes. Semin Cell Dev Biol 2016; 51:106-16. [PMID: 26806634 DOI: 10.1016/j.semcdb.2016.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 01/06/2016] [Indexed: 02/07/2023]
Abstract
Diabetes is a chronic disease caused by the loss or dysfunction of the insulin-producing β-cells in the pancreas. To date, much of our knowledge about β-cells in humans comes from studying rare monogenic forms of diabetes. Importantly, the majority of mutations so far associated to monogenic diabetes are in genes that exert a regulatory role in pancreatic development and/or β-cell function. Thus, the identification and study of novel mutations open an unprecedented window into human pancreatic development. In this review, we summarize major advances in the genetic dissection of different types of monogenic diabetes and the insights gained from a developmental perspective. We highlight future challenges to bridge the gap between the fast accumulation of genetic data through next-generation sequencing and the need of functional insights into disease mechanisms. Lastly, we discuss the relevance and advantages of studying candidate gene variants in vivo using the Xenopus as model system.
Collapse
Affiliation(s)
- Julia Kofent
- Max Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, D-13125 Berlin, Germany
| | - Francesca M Spagnoli
- Max Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, D-13125 Berlin, Germany.
| |
Collapse
|
33
|
Transplantation of mesenchymal stem cells improves type 1 diabetes mellitus. Cell Tissue Res 2015; 364:345-55. [PMID: 26650464 DOI: 10.1007/s00441-015-2330-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 11/06/2015] [Indexed: 12/29/2022]
Abstract
Bone-marrow-derived stem cells can regenerate pancreatic tissue in a model of type 1 diabetes mellitus. Mesenchymal stem cells (MSCs) form the main part of bone marrow. We show that the intrapancreatic transplantation of MSCs elevates serum insulin and C-peptide, while decreasing blood glucose. MSCs engrafted into the damaged rat pancreas become distributed into the blood vessels, acini, ducts, and islets. Renascent islets, islet-like clusters, and a small number of MSCs expressing insulin protein have been observed in the pancreas of diabetic rats. Intrapancreatic transplantation of MSCs triggers a series of molecular and cellular events, including differentiation towards the pancreas directly and the provision of a niche to start endogenous pancreatic regeneration, which ameliorates hypoinsulinemia and hyperglycemia caused by streptozotocin. These data establish the many roles of MSCs in the restoration of the function of an injured organ.
Collapse
|
34
|
Co-culture with mature islet cells augments the differentiation of insulin-producing cells from pluripotent stem cells. Stem Cell Rev Rep 2015; 11:62-74. [PMID: 25173880 DOI: 10.1007/s12015-014-9554-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Islet transplantation has been hampered by the shortage of islet donors available for diabetes therapy. However, pluripotent stem cells (PSCs) can be an alternative source of insulin-producing cells (IPCs) because of their capacity for self-renewal and differentiation. We described a method to efficiently differentiate PSCs into IPCs by co-culturing mature islets with directed-differentiated pancreatic endoderm (PE) cells from mouse and human PSCs. PE cells co-cultured with islet cells or islet cell-derived conditioned medium (CM) showed increased expression levels of β-cell markers; significantly higher levels of proinsulin- and Newport Green (NG)-positive cells, which revealed the characteristics of insulin producing cells; and increased insulin secretion upon glucose stimulation. Co-culturing human PE cells with islet cells was also effective to differentiate PE cells into IPCs. Diabetic nude mice transplanted with co-cultured cells exhibited restored euglycemia, human C-peptide release, and improved glucose tolerance. Immunohistochemistry revealed that insulin+/C-peptide + cells existed in the grafted tissues. These results suggest that mature islet cells can increase the differentiation efficiency of PE cells into mature IPCs via paracrine effects.
Collapse
|
35
|
Larsen BM, Hrycaj SM, Newman M, Li Y, Wellik DM. Mesenchymal Hox6 function is required for mouse pancreatic endocrine cell differentiation. Development 2015; 142:3859-68. [PMID: 26450967 DOI: 10.1242/dev.126888] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/30/2015] [Indexed: 12/20/2022]
Abstract
Despite significant advances in our understanding of pancreatic endocrine cell development, the function of the pancreatic mesodermal niche in this process is poorly understood. Here we report a novel role for mouse Hox6 genes in pancreatic organogenesis. Hox6 genes are expressed exclusively in the mesoderm of the developing pancreas. Genetic loss of all three Hox6 paralogs (Hoxa6, Hoxb6 and Hoxc6) leads to a dramatic loss of endoderm-derived endocrine cells, including insulin-secreting β-cells, and to mild delays and disruptions in pancreatic branching and exocrine differentiation. Ngn3-expressing pan-endocrine progenitor cells are specified normally in Hox6 mutant pancreata, but fail to mature into hormone-producing cells. Reduced expression of Wnt5a is observed in mutant pancreatic mesenchyme, leading to subsequent loss of expression of the crucial Wnt inhibitors Sfrp3 and Dkk1 in endocrine progenitor cells. These results reveal a key role for Hox6 genes in establishing Wnt mesenchymal-epithelial crosstalk in pancreatic development.
Collapse
Affiliation(s)
- Brian M Larsen
- Department of Internal Medicine, Division of Molecular Medicine and Genetics, University of Michigan, Ann Arbor, MI 48109-2200, USA Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Steven M Hrycaj
- Department of Internal Medicine, Division of Molecular Medicine and Genetics, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Micaleah Newman
- Department of Internal Medicine, Division of Molecular Medicine and Genetics, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Ye Li
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Deneen M Wellik
- Department of Internal Medicine, Division of Molecular Medicine and Genetics, University of Michigan, Ann Arbor, MI 48109-2200, USA Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109-2200, USA Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| |
Collapse
|
36
|
Bonfanti P, Nobecourt E, Oshima M, Albagli-Curiel O, Laurysens V, Stangé G, Sojoodi M, Heremans Y, Heimberg H, Scharfmann R. Ex Vivo Expansion and Differentiation of Human and Mouse Fetal Pancreatic Progenitors Are Modulated by Epidermal Growth Factor. Stem Cells Dev 2015; 24:1766-78. [DOI: 10.1089/scd.2014.0550] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Paola Bonfanti
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Estelle Nobecourt
- INSERM, U1016, Institut Cochin, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris, Paris, France
| | - Masaya Oshima
- INSERM, U1016, Institut Cochin, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris, Paris, France
| | - Olivier Albagli-Curiel
- INSERM, U1016, Institut Cochin, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris, Paris, France
| | - Veerle Laurysens
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Geert Stangé
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mozhdeh Sojoodi
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yves Heremans
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Harry Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Raphael Scharfmann
- INSERM, U1016, Institut Cochin, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris, Paris, France
| |
Collapse
|
37
|
Alejandro EU, Gregg B, Blandino-Rosano M, Cras-Méneur C, Bernal-Mizrachi E. Natural history of β-cell adaptation and failure in type 2 diabetes. Mol Aspects Med 2014; 42:19-41. [PMID: 25542976 DOI: 10.1016/j.mam.2014.12.002] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 11/04/2014] [Accepted: 12/05/2014] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes mellitus (T2D) is a complex disease characterized by β-cell failure in the setting of insulin resistance. The current evidence suggests that genetic predisposition, and environmental factors can impair the capacity of the β-cells to respond to insulin resistance and ultimately lead to their failure. However, genetic studies have demonstrated that known variants account for less than 10% of the overall estimated T2D risk, suggesting that additional unidentified factors contribute to susceptibility of this disease. In this review, we will discuss the different stages that contribute to the development of β-cell failure in T2D. We divide the natural history of this process in three major stages: susceptibility, β-cell adaptation and β-cell failure, and provide an overview of the molecular mechanisms involved. Further research into mechanisms will reveal key modulators of β-cell failure and thus identify possible novel therapeutic targets and potential interventions to protect against β-cell failure.
Collapse
Affiliation(s)
- Emilyn U Alejandro
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA
| | - Brigid Gregg
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Manuel Blandino-Rosano
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA
| | - Corentin Cras-Méneur
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA
| | - Ernesto Bernal-Mizrachi
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA; VA Ann Arbor Healthcare System, Ann Arbor, MI, USA.
| |
Collapse
|
38
|
Kumar SS, Alarfaj AA, Munusamy MA, Singh AJAR, Peng IC, Priya SP, Hamat RA, Higuchi A. Recent developments in β-cell differentiation of pluripotent stem cells induced by small and large molecules. Int J Mol Sci 2014; 15:23418-47. [PMID: 25526563 PMCID: PMC4284775 DOI: 10.3390/ijms151223418] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 12/03/2014] [Accepted: 12/08/2014] [Indexed: 12/21/2022] Open
Abstract
Human pluripotent stem cells, including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), hold promise as novel therapeutic tools for diabetes treatment because of their self-renewal capacity and ability to differentiate into beta (β)-cells. Small and large molecules play important roles in each stage of β-cell differentiation from both hESCs and hiPSCs. The small and large molecules that are described in this review have significantly advanced efforts to cure diabetic disease. Lately, effective protocols have been implemented to induce hESCs and human mesenchymal stem cells (hMSCs) to differentiate into functional β-cells. Several small molecules, proteins, and growth factors promote pancreatic differentiation from hESCs and hMSCs. These small molecules (e.g., cyclopamine, wortmannin, retinoic acid, and sodium butyrate) and large molecules (e.g. activin A, betacellulin, bone morphogentic protein (BMP4), epidermal growth factor (EGF), fibroblast growth factor (FGF), keratinocyte growth factor (KGF), hepatocyte growth factor (HGF), noggin, transforming growth factor (TGF-α), and WNT3A) are thought to contribute from the initial stages of definitive endoderm formation to the final stages of maturation of functional endocrine cells. We discuss the importance of such small and large molecules in uniquely optimized protocols of β-cell differentiation from stem cells. A global understanding of various small and large molecules and their functions will help to establish an efficient protocol for β-cell differentiation.
Collapse
Affiliation(s)
- S Suresh Kumar
- Department of Medical Microbiology and Parasitology, Universities Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Abdullah A Alarfaj
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Murugan A Munusamy
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - A J A Ranjith Singh
- Department of Bioscience, Jacintha Peter College of Arts and Sciences, Ayakudi, Tenkasi, Tamilnadu 627852, India.
| | - I-Chia Peng
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan 32001, Taiwan.
| | - Sivan Padma Priya
- Department of Basic Science and Department of Surgical Sciences, Ajman University of Science and Technology-Fujairah Campus, P.O. Box 9520, Al Fujairah, United Arab Emirates.
| | - Rukman Awang Hamat
- Department of Medical Microbiology and Parasitology, Universities Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Akon Higuchi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
39
|
Gregg B, Elghazi L, Alejandro EU, Smith MR, Blandino-Rosano M, El-Gabri D, Cras-Méneur C, Bernal-Mizrachi E. Exposure of mouse embryonic pancreas to metformin enhances the number of pancreatic progenitors. Diabetologia 2014; 57:2566-75. [PMID: 25249235 PMCID: PMC4417192 DOI: 10.1007/s00125-014-3379-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 08/28/2014] [Indexed: 01/23/2023]
Abstract
AIMS/HYPOTHESIS Developing beta cells are vulnerable to nutrient environmental signals. Early developmental processes that alter the number of pancreatic progenitors can determine the number of beta cells present at birth. Metformin, the most widely used oral agent for treating diabetes, alters intracellular energy status in part by increasing AMP-activated protein kinase (AMPK) signalling. This study examined the effect of metformin on developing pancreas and beta cells. METHODS Pancreatic rudiments from CD-1 mice at embryonic day 13.0 (E13.0) were cultured with metformin, 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR, an AMPK activator) or vehicle control in vitro. In another set of studies, pregnant C57BL/6 mice were treated with metformin throughout gestation. Embryonic (E14.0) and neonatal pancreases were then analysed for their morphometry. RESULTS In vitro metformin treatment led to an increase in the proliferation and number of pancreatic duodenal homeobox 1-positive (PDX1(+)) progenitors. These results were reproduced by in vitro culture of embryonic pancreas rudiments with AICAR, suggesting that AMPK activation was involved. Similarly, metformin administration to pregnant dams induced an increase in both PDX1(+) and neurogenin 3-positive progenitors in the embryonic pancreas at E14.0 and these changes resulted in an increased beta cell fraction in neonates. CONCLUSIONS/INTERPRETATION These results indicate that exposure to metformin during gestation modulates the early steps of beta cell development (prior to E14.0) towards an increase in the number of pancreatic and endocrine progenitors. These changes ultimately result in a higher beta cell fraction at birth. These findings are of clinical importance given that metformin is currently used for the treatment of gestational diabetes.
Collapse
Affiliation(s)
- Brigid Gregg
- Department of Pediatrics, Division of Endocrinology, Diabetes and Metabolism, University of Michigan, Ann Arbor, Michigan, USA
| | - Lynda Elghazi
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Emilyn U. Alejandro
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Michelle R. Smith
- Department of Pediatrics, Division of Endocrinology, Diabetes and Metabolism, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Manuel Blandino-Rosano
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Deena El-Gabri
- Department of Pediatrics, Division of Endocrinology, Diabetes and Metabolism, University of Michigan, Ann Arbor, Michigan, USA
| | - Corentin Cras-Méneur
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Ernesto Bernal-Mizrachi
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, Michigan, USA
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan
| |
Collapse
|
40
|
Vázquez P, Robles AM, de Pablo F, Hernández-Sánchez C. Non-neural tyrosine hydroxylase, via modulation of endocrine pancreatic precursors, is required for normal development of beta cells in the mouse pancreas. Diabetologia 2014; 57:2339-47. [PMID: 25082160 PMCID: PMC4181516 DOI: 10.1007/s00125-014-3341-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 07/01/2014] [Indexed: 11/07/2022]
Abstract
AIMS/HYPOTHESIS Apart from transcription factors, little is known about the molecules that modulate the proliferation and differentiation of pancreatic endocrine cells. The early expression of tyrosine hydroxylase (TH) in a subset of glucagon(+) cells led us to investigate whether catecholamines have a role in beta cell development. METHODS We studied the immunohistochemical characteristics of TH-expressing cells in wild-type (Th (+/+) ) mice during early pancreas development, and analysed the endocrine pancreas phenotype of TH-deficient (Th (-/-) ) mice. We also studied the effect of dopamine addition and TH-inhibition on insulin-producing cells in explant cultures. RESULTS In the mouse pancreas at embryonic day (E)12.5-E13.5, the ∼10% of early glucagon(+) cells that co-expressed TH rarely proliferated and did not express the precursor marker neurogenin 3 at E13.5. The number of insulin(+) cells in the Th (-/-) embryonic pancreas was decreased as compared with wild-type embryos at E13.5. While no changes in pancreatic and duodenal homeobox 1 (PDX1)(+)-progenitor cell number were observed between groups at E12.5, the number of neurogenin 3 and NK2 homeobox 2 (NKX2.2)-expressing cells was reduced in Th (-/-) embryonic pancreas, an effect that occurred in parallel with increased expression of the transcriptional repressor Hes1. The potential role of dopamine as a pro-beta cell stimulus was tested by treating pancreas explants with this catecholamine, which resulted in an increase in total insulin content and insulin(+) cells relative to control explants. CONCLUSIONS/INTERPRETATION A non-neural catecholaminergic pathway appears to modulate the pancreatic endocrine precursor and insulin producing cell neogenesis. This finding may have important implications for approaches seeking to promote the generation of beta cells to treat diabetes.
Collapse
Affiliation(s)
- Patricia Vázquez
- 3D (Development, Differentiation, Degeneration) Lab, Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (ISCIII), Ministerio de Economía y Competitividad, Spain, http://www.ciberdem.org/
| | - Ana M. Robles
- 3D (Development, Differentiation, Degeneration) Lab, Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Flora de Pablo
- 3D (Development, Differentiation, Degeneration) Lab, Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (ISCIII), Ministerio de Economía y Competitividad, Spain, http://www.ciberdem.org/
| | - Catalina Hernández-Sánchez
- 3D (Development, Differentiation, Degeneration) Lab, Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (ISCIII), Ministerio de Economía y Competitividad, Spain, http://www.ciberdem.org/
| |
Collapse
|
41
|
Soggia A, Ramond C, Akiyama H, Scharfmann R, Duvillie B. von Hippel-Lindau gene disruption in mouse pancreatic progenitors and its consequences on endocrine differentiation in vivo: importance of HIF1-α and VEGF-A upregulation. Diabetologia 2014; 57:2348-56. [PMID: 25186293 DOI: 10.1007/s00125-014-3365-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 08/11/2014] [Indexed: 12/15/2022]
Abstract
AIM/HYPOTHESIS Different studies have linked hypoxia to embryonic development. Specifically, when embryonic pancreases are cultured ex vivo under hypoxic conditions (3% O2), beta cell development is impaired. Different cellular signalling pathways are involved in adaptation to hypoxia, including the ubiquitous hypoxia-inducible-factor 1-α (HIF1-α) pathway. We aimed to analyse the effects of HIF1-α stabilisation on fetal pancreas development in vivo. METHODS We deleted the Vhl gene, which encodes von Hippel-Lindau protein (pVHL), a factor necessary for HIF1-α degradation, by crossing Vhl-floxed mice with Sox9-Cre mice. RESULTS HIF1-α was stabilised in pancreatic progenitor cells in which the HIF pathway was induced. The number of neurogenin-3 (NGN3)-expressing cells was reduced and consequently endocrine development was altered in Vhl knockout pancreases. HIF1-α stabilisation induced Vegfa upregulation, leading to increased vascularisation. To investigate the impact of increased vascularisation on NGN3 expression, we used a bioassay in which Vhl mutant pancreases were cultured with or without vascular endothelial growth factor (VEGF) receptor 2 (VEGF-R2) inhibitors (e.g. Ki8751). Ex vivo analysis showed that Vhl knockout pancreases developed fewer NGN3-positive cells compared with controls. Interestingly, this effect was blocked when vascularisation was inhibited in the presence of VEGF-R2 inhibitors. CONCLUSIONS/INTERPRETATION Our data demonstrate that HIF1-α negatively controls beta cell differentiation in vivo by regulating NGN3 expression, and that this effect is mediated by signals from blood vessels.
Collapse
Affiliation(s)
- Andrea Soggia
- U1016 Inserm/Institut Cochin, Groupe Hospitalier Cochin Port-Royal, Bâtiment Cassini, 123 Boulevard du Port-Royal, 75014, Paris, France
| | | | | | | | | |
Collapse
|
42
|
Hoarau E, Chandra V, Rustin P, Scharfmann R, Duvillie B. Pro-oxidant/antioxidant balance controls pancreatic β-cell differentiation through the ERK1/2 pathway. Cell Death Dis 2014; 5:e1487. [PMID: 25341041 PMCID: PMC4237262 DOI: 10.1038/cddis.2014.441] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 09/06/2014] [Accepted: 09/08/2014] [Indexed: 12/20/2022]
Abstract
During embryogenesis, the intrauterine milieu affects cell proliferation, differentiation, and function by modifying gene expression in susceptible cells, such as the pancreatic β-cells. In this limited energy environment, mitochondrial dysfunction can lead to overproduction of reactive oxygen species (ROS) and to a decline in β-cell function. In opposition to this toxicity, ROS are also required for insulin secretion. Here we investigated the role of ROS in β-cell development. Surprisingly, decreasing ROS production in vivo reduced β-cell differentiation. Moreover, in cultures of pancreatic explants, progenitors were highly sensitive to ROS stimulation and responded by generating β-cells. ROS enhanced β-cell differentiation through modulation of ERK1/2 signaling. Gene transfer and pharmacological manipulations, which diminish cellular ROS levels, also interfered with normal β-cell differentiation. This study highlights the role of the redox balance on β-cell development and provides information that will be useful for improving β-cell production from embryonic stem cells, a step in cell therapy for diabetes.
Collapse
Affiliation(s)
- E Hoarau
- 1] INSERM, U1016, Institut Cochin, Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - V Chandra
- 1] INSERM, U1016, Institut Cochin, Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - P Rustin
- INSERM U676, Hopital Robert Debre, Paris, France
| | - R Scharfmann
- 1] INSERM, U1016, Institut Cochin, Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - B Duvillie
- 1] INSERM, U1016, Institut Cochin, Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| |
Collapse
|
43
|
Greggio C, De Franceschi F, Figueiredo-Larsen M, Grapin-Botton A. In vitro pancreas organogenesis from dispersed mouse embryonic progenitors. J Vis Exp 2014. [PMID: 25079453 DOI: 10.3791/51725] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The pancreas is an essential organ that regulates glucose homeostasis and secretes digestive enzymes. Research on pancreas embryogenesis has led to the development of protocols to produce pancreatic cells from stem cells (1). The whole embryonic organ can be cultured at multiple stages of development (2-4). These culture methods have been useful to test drugs and to image developmental processes. However the expansion of the organ is very limited and morphogenesis is not faithfully recapitulated since the organ flattens. We propose three-dimensional (3D) culture conditions that enable the efficient expansion of dissociated mouse embryonic pancreatic progenitors. By manipulating the composition of the culture medium it is possible to generate either hollow spheres, mainly composed of pancreatic progenitors expanding in their initial state, or, complex organoids which progress to more mature expanding progenitors and differentiate into endocrine, acinar and ductal cells and which spontaneously self-organize to resemble the embryonic pancreas. We show here that the in vitro process recapitulates many aspects of natural pancreas development. This culture system is suitable to investigate how cells cooperate to form an organ by reducing its initial complexity to few progenitors. It is a model that reproduces the 3D architecture of the pancreas and that is therefore useful to study morphogenesis, including polarization of epithelial structures and branching. It is also appropriate to assess the response to mechanical cues of the niche such as stiffness and the effects on cell´s tensegrity.
Collapse
Affiliation(s)
- Chiara Greggio
- Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, Swiss Institute for Experimental Cancer Research
| | - Filippo De Franceschi
- Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, Swiss Institute for Experimental Cancer Research
| | | | - Anne Grapin-Botton
- Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, Swiss Institute for Experimental Cancer Research; DanStem, University of Copenhagen;
| |
Collapse
|
44
|
Li X, Nadauld L, Ootani A, Corney DC, Pai RK, Gevaert O, Cantrell MA, Rack PG, Neal JT, Chan CWM, Yeung T, Gong X, Yuan J, Wilhelmy J, Robine S, Attardi LD, Plevritis SK, Hung KE, Chen CZ, Ji HP, Kuo CJ. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat Med 2014; 20:769-77. [PMID: 24859528 PMCID: PMC4087144 DOI: 10.1038/nm.3585] [Citation(s) in RCA: 343] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 01/29/2014] [Indexed: 01/10/2023]
Abstract
The application of primary organoid cultures containing epithelial and mesenchymal elements to cancer modeling holds promise for combining the accurate multilineage differentiation and physiology of in vivo systems with the facile in vitro manipulation of transformed cell lines. Here we used a single air-liquid interface culture method without modification to engineer oncogenic mutations into primary epithelial and mesenchymal organoids from mouse colon, stomach and pancreas. Pancreatic and gastric organoids exhibited dysplasia as a result of expression of Kras carrying the G12D mutation (Kras(G12D)), p53 loss or both and readily generated adenocarcinoma after in vivo transplantation. In contrast, primary colon organoids required combinatorial Apc, p53, Kras(G12D) and Smad4 mutations for progressive transformation to invasive adenocarcinoma-like histology in vitro and tumorigenicity in vivo, recapitulating multi-hit models of colorectal cancer (CRC), as compared to the more promiscuous transformation of small intestinal organoids. Colon organoid culture functionally validated the microRNA miR-483 as a dominant driver oncogene at the IGF2 (insulin-like growth factor-2) 11p15.5 CRC amplicon, inducing dysplasia in vitro and tumorigenicity in vivo. These studies demonstrate the general utility of a highly tractable primary organoid system for cancer modeling and driver oncogene validation in diverse gastrointestinal tissues.
Collapse
Affiliation(s)
- Xingnan Li
- Department of Medicine, Hematology Division, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Lincoln Nadauld
- Department of Medicine, Hematology Division, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Akifumi Ootani
- Department of Medicine, Hematology Division, Stanford University School of Medicine, Stanford, CA 94305 USA
- Department of Internal Medicine, Saga Medical School, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - David C. Corney
- Department of Medicine, Hematology Division, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Reetesh K. Pai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Olivier Gevaert
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Michael A. Cantrell
- Department of Medicine, Hematology Division, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Paul G. Rack
- Department of Medicine, Hematology Division, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - James T. Neal
- Department of Medicine, Hematology Division, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Carol W-M. Chan
- Department of Medicine, Hematology Division, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Trevor Yeung
- Department of Medicine, Hematology Division, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Xue Gong
- Baxter Laboratories and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Jenny Yuan
- Department of Medicine, Hematology Division, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Julie Wilhelmy
- Division of Oncology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Sylvie Robine
- Equipe de Morphogenèse et Signalisation cellulaires, UMR 144 CNRS/Institut Curie, 26 rue d’Ulm, 75248 Paris cedex 05, France
| | - Laura D. Attardi
- Division of Radiation Biology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Sylvia K. Plevritis
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Kenneth E. Hung
- Department of Medicine, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Chang-Zheng Chen
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Hanlee P. Ji
- Division of Oncology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Calvin J. Kuo
- Department of Medicine, Hematology Division, Stanford University School of Medicine, Stanford, CA 94305 USA
| |
Collapse
|
45
|
Schiesser JV, Micallef SJ, Hawes S, Elefanty AG, Stanley EG. Derivation of insulin-producing beta-cells from human pluripotent stem cells. Rev Diabet Stud 2014; 11:6-18. [PMID: 25148364 DOI: 10.1900/rds.2014.11.6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Human embryonic stem cells have been advanced as a source of insulin-producing cells that could potentially replace cadaveric-derived islets in the treatment of type 1 diabetes. To this end, protocols have been developed that promote the formation of pancreatic progenitors and endocrine cells from human pluripotent stem cells, encompassing both embryonic stem cells and induced pluripotent stem cells. In this review, we examine these methods and place them in the context of the developmental and embryological studies upon which they are based. In particular, we outline the stepwise differentiation of cells towards definitive endoderm, pancreatic endoderm, endocrine lineages and the emergence of functional beta-cells. In doing so, we identify key factors common to many such protocols and discuss the proposed action of these factors in the context of cellular differentiation and ongoing development. We also compare strategies that entail transplantation of progenitor populations with those that seek to develop fully functional hormone expressing cells in vitro. Overall, our survey of the literature highlights the significant progress already made in the field and identifies remaining deficiencies in developing a pluripotent stem cell based treatment for type 1 diabetes.
Collapse
Affiliation(s)
- Jacqueline V Schiesser
- Monash Immunology and Stem Cell Laboratories (MISCL), Level 3, Building 75, STRIP1, West Ring Road, Monash University, Clayton, Victoria, 3800, Australia
| | - Suzanne J Micallef
- Monash Immunology and Stem Cell Laboratories (MISCL), Level 3, Building 75, STRIP1, West Ring Road, Monash University, Clayton, Victoria, 3800, Australia
| | - Susan Hawes
- Monash Immunology and Stem Cell Laboratories (MISCL), Level 3, Building 75, STRIP1, West Ring Road, Monash University, Clayton, Victoria, 3800, Australia
| | - Andrew G Elefanty
- Monash Immunology and Stem Cell Laboratories (MISCL), Level 3, Building 75, STRIP1, West Ring Road, Monash University, Clayton, Victoria, 3800, Australia
| | - Edouard G Stanley
- Monash Immunology and Stem Cell Laboratories (MISCL), Level 3, Building 75, STRIP1, West Ring Road, Monash University, Clayton, Victoria, 3800, Australia
| |
Collapse
|
46
|
Rachdi L, Kariyawasam D, Guez F, Aïello V, Arbonés ML, Janel N, Delabar JM, Polak M, Scharfmann R. Dyrk1a haploinsufficiency induces diabetes in mice through decreased pancreatic beta cell mass. Diabetologia 2014; 57:960-9. [PMID: 24477974 DOI: 10.1007/s00125-014-3174-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 01/06/2014] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS Growth factors and nutrients are important regulators of pancreatic beta cell mass and function. However, the signalling pathways by which these factors modulate these processes have not yet been fully elucidated. DYRK1A (also named minibrain/MNB) is a member of the dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) family that has been conserved across evolution. A significant amount of data implicates DYRK1A in brain growth and function, as well as in neurodegenerative processes in Alzheimer's disease and Down's syndrome. We investigated here whether DYRK1A would be an attractive candidate for beta cell growth modulation. METHODS To study the role of DYRK1A in beta cell growth, we used Dyrk1a-deficient mice. RESULTS We show that DYRK1A is expressed in pancreatic islets and provide evidence that changes in Dyrk1a gene dosage in mice strongly modulate glycaemia and circulating insulin levels. Specifically, Dyrk1a-haploinsufficient mice show severe glucose intolerance, reduced beta cell mass and decreased beta cell proliferation. CONCLUSIONS/INTERPRETATION Taken together, our data indicate that DYRK1A is a critical kinase for beta cell growth as Dyrk1a-haploinsufficient mice show a diabetic profile.
Collapse
Affiliation(s)
- Latif Rachdi
- INSERM U1016, Institut Cochin, Faculté de Médecine Cochin, Université Paris Descartes, 24 Rue du Faubourg St Jacques, 75014, Paris, France,
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Aïello V, Moreno-Asso A, Servitja JM, Martín M. Thyroid hormones promote endocrine differentiation at expenses of exocrine tissue. Exp Cell Res 2014; 322:236-48. [DOI: 10.1016/j.yexcr.2014.01.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 01/17/2014] [Accepted: 01/27/2014] [Indexed: 12/12/2022]
|
48
|
Capito C, Simon MT, Aiello V, Clark A, Aigrain Y, Ravassard P, Scharfmann R. Mouse muscle as an ectopic permissive site for human pancreatic development. Diabetes 2013; 62:3479-87. [PMID: 23835344 PMCID: PMC3781474 DOI: 10.2337/db13-0554] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
While sporadic human genetic studies have permitted some comparisons between rodent and human pancreatic development, the lack of a robust experimental system has not permitted detailed examination of human pancreatic development. We previously developed a xenograft model of immature human fetal pancreas grafted under the kidney capsule of immune-incompetent mice, which allowed the development of human pancreatic β-cells. Here, we compared the development of human and murine fetal pancreatic grafts either under skeletal muscle epimysium or under the renal capsule. We demonstrated that human pancreatic β-cell development occurs more slowly (weeks) than murine pancreas (days) both by differentiation of pancreatic progenitors and by proliferation of developing β-cells. The superficial location of the skeletal muscle graft and its easier access permitted in vivo lentivirus-mediated gene transfer with a green fluorescent protein-labeled construct under control of the insulin or elastase gene promoter, which targeted β-cells and nonendocrine cells, respectively. This model of engraftment under the skeletal muscle epimysium is a new approach for longitudinal studies, which allows localized manipulation to determine the regulation of human pancreatic development.
Collapse
Affiliation(s)
- Carmen Capito
- INSERM U845, Research Center Growth and Signalling, Faculté de Médecine Cochin, Université Paris Descartes, Paris, France
| | - Marie-Thérèse Simon
- INSERM U845, Research Center Growth and Signalling, Faculté de Médecine Cochin, Université Paris Descartes, Paris, France
| | - Virginie Aiello
- INSERM U845, Research Center Growth and Signalling, Faculté de Médecine Cochin, Université Paris Descartes, Paris, France
| | - Anne Clark
- Diabetes Research Laboratories, Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, U.K
| | - Yves Aigrain
- Necker Enfants Malades University Hospital, Université Paris Descartes, Paris, France
| | - Philippe Ravassard
- Biotechnology and Biotherapy Team, Université Pierre et Marie Curie-Paris 6, Biotechnology and Biotherapy Team, Centre de Recherche de l’Institut du Cerveau et de la Moelle épinière, UMRS 975, CNRS, UMR 7225, INSERM U975, Paris, France
| | - Raphael Scharfmann
- INSERM U845, Research Center Growth and Signalling, Faculté de Médecine Cochin, Université Paris Descartes, Paris, France
- Corresponding author: Raphael Scharfmann,
| |
Collapse
|
49
|
Duvillié B. Quelles cellules souches pour une réparation du pancréas endocrine ? Med Sci (Paris) 2013; 29:744-8. [DOI: 10.1051/medsci/2013298013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
50
|
Abstract
PURPOSE OF REVIEW This review evaluates recent progress in several approaches aimed at developing human surrogate β cells, and identifies gaps that need to be filled for bringing them closer to clinical application. RECENT FINDINGS Cells expanded in vitro from human cadaver donor β cells under conditions causing dedifferentiation have been shown to undergo redifferentiation following inhibition of the Notch pathway. Efforts for differentiation of insulin-producing cells from human pluripotent stem cells have focused on isolation and expansion of intermediate-stage cells. The role of mesenchyme in expansion of pancreas progenitors has been emphasized by mouse cell ablation, and co-culture of human embryonic stem cell-derived definitive endoderm with mesenchyme. Incomplete removal of Polycomb-mediated repression of endocrine genes in embryonic stem cell-derived insulin-producing cells generated in vitro has been suggested to be responsible for their immature phenotype. Induced pluripotent stem cells reprogrammed from β cells have been shown to exhibit an enhanced differentiation capacity toward insulin-producing cells, compared with other pluripotent stem cells. A new approach for reprogramming non-β into β-like cells involving transcription factor gene ablation has been demonstrated in mouse enteroendocrine cells in vivo. SUMMARY New insights into the stumbling blocks in expansion of human donor islet cells, differentiation of pluripotent stem cells, and reprogramming of non-β cell types are shaping improved strategies, which are likely to bring us closer to the goal of generating abundant human surrogate β cells.
Collapse
Affiliation(s)
- Shimon Efrat
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University Ramat Aviv, Tel Aviv, Israel.
| |
Collapse
|