1
|
Lam T, Quach HT, Hall L, Abou Chakra M, Wong AP. A multidisciplinary approach towards modeling of a virtual human lung. NPJ Syst Biol Appl 2025; 11:38. [PMID: 40251169 PMCID: PMC12008392 DOI: 10.1038/s41540-025-00517-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
Integrating biological data with in silico modeling offers the transformative potential to develop virtual human models, or "digital twins." These models hold immense promise for deepening our understanding of diseases and uncovering new therapeutic strategies. This approach is especially valuable for diseases lacking reliable models. Here we review current modelling efforts in of human lung development, highlighting the role of interdisciplinary collaboration and key advances toward a digital lung twin.
Collapse
Affiliation(s)
- Timothy Lam
- Program in Developmental, Stem cell and Cancer Biology, Hospital for Sick Children, PGCRL 16-9420, Toronto, ON, Canada
| | - Henry T Quach
- Program in Developmental, Stem cell and Cancer Biology, Hospital for Sick Children, PGCRL 16-9420, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Lauren Hall
- Program in Developmental, Stem cell and Cancer Biology, Hospital for Sick Children, PGCRL 16-9420, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Maria Abou Chakra
- Donnelly Centre for Cellular and Biomedical Research, University of Toronto, Toronto, ON, Canada
| | - Amy P Wong
- Program in Developmental, Stem cell and Cancer Biology, Hospital for Sick Children, PGCRL 16-9420, Toronto, ON, Canada.
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
2
|
Li Y, Zhu J, Yue C, Song S, Tian L, Wang Y. Recent advances in pancreatic α-cell transdifferentiation for diabetes therapy. Front Immunol 2025; 16:1551372. [PMID: 39911402 PMCID: PMC11794509 DOI: 10.3389/fimmu.2025.1551372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
As the global prevalence of diabetes mellitus rises, traditional treatments like insulin therapy and oral hypoglycemic agents often fail to achieve optimal glycemic control, leading to severe complications. Recent research has focused on replenishing pancreatic β-cells through the transdifferentiation of α-cells, offering a promising therapeutic avenue. This review explores the molecular mechanisms underlying α-cell to β-cell transdifferentiation, emphasizing key transcription factors such as Dnmt1, Arx, Pdx1, MafA, and Nkx6.1. The potential clinical applications, especially in type 1 and type 2 diabetes characterized by significant β-cell dysfunction, are addressed. Challenges, including low transdifferentiation efficiency, cell stability, and safety concerns, are also included. Future research directions include optimizing molecular pathways, enhancing transdifferentiation efficiency, and ensuring the long-term stability of β-cell identity. Overall, the ability to convert α-cells into β-cells represents a transformative strategy for diabetes treatment, offering hope for more effective and sustainable therapies for patients with severe β-cell loss.
Collapse
Affiliation(s)
- Yanjiao Li
- Department of Pharmacy, Qionglai Hospital of Traditional Chinese Medicine, Chengdu, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jinyu Zhu
- Center for Geriatrics and Endocrinology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Congyang Yue
- Center for Geriatrics and Endocrinology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Limin Tian
- Center for Geriatrics and Endocrinology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yi Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Center for Geriatrics and Endocrinology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Center for Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Wang L, Baek S, Prasad G, Wildenthal J, Guo K, Sturgill D, Truongvo T, Char E, Pegoraro G, McKinnon K, Hoskins JW, Amundadottir LT, Arda HE. Predictive Prioritization of Enhancers Associated with Pancreas Disease Risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.07.611794. [PMID: 39314336 PMCID: PMC11418953 DOI: 10.1101/2024.09.07.611794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Genetic and epigenetic variations in regulatory enhancer elements increase susceptibility to a range of pathologies. Despite recent advances, linking enhancer elements to target genes and predicting transcriptional outcomes of enhancer dysfunction remain significant challenges. Using 3D chromatin conformation assays, we generated an extensive enhancer interaction dataset for the human pancreas, encompassing more than 20 donors and five major cell types, including both exocrine and endocrine compartments. We employed a network approach to parse chromatin interactions into enhancer-promoter tree models, facilitating a quantitative, genome-wide analysis of enhancer connectivity. With these tree models, we developed a machine learning algorithm to estimate the impact of enhancer perturbations on cell type-specific gene expression in the human pancreas. Orthogonal to our computational approach, we perturbed enhancer function in primary human pancreas cells using CRISPR interference and quantified the effects at the single-cell level through RNA FISH coupled with high-throughput imaging. Our enhancer tree models enabled the annotation of common germline risk variants associated with pancreas diseases, linking them to putative target genes in specific cell types. For pancreatic ductal adenocarcinoma, we found a stronger enrichment of disease susceptibility variants within acinar cell regulatory elements, despite ductal cells historically being assumed as the primary cell-of-origin. Our integrative approach-combining cell type-specific enhancer-promoter interaction mapping, computational models, and single-cell enhancer perturbation assays-produced a robust resource for studying the genetic basis of pancreas disorders.
Collapse
Affiliation(s)
- Li Wang
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Songjoon Baek
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gauri Prasad
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Translational Genomics, Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John Wildenthal
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Konnie Guo
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Sturgill
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thucnhi Truongvo
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Erin Char
- Laboratory of Translational Genomics, Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gianluca Pegoraro
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katherine McKinnon
- Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | - Jason W. Hoskins
- Laboratory of Translational Genomics, Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laufey T. Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - H. Efsun Arda
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
4
|
Imamura H, Adachi T, Miyamoto D, Kin T, Yamashita M, Matsushima H, Hara T, Soyama A, Eguchi S. Analysis of Expression Status and Relationship Between Senescence-related Genes and Pancreatic Function-related Genes in Human Islets of Langerhans from Donors of Various Ages. In Vivo 2024; 38:2165-2171. [PMID: 39187349 PMCID: PMC11363790 DOI: 10.21873/invivo.13679] [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: 04/19/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND/AIM Although studies on senescence-related genes using human islets of Langerhans have been performed, the expression of senescence-related genes and their association with functional genes in islets remain insufficiently investigated. We aimed to determine whether and what types of senescent-related genes are expressed in islets and identify their correlations with pancreatic function-related genes by using islets isolated for transplantation from individuals of various ages. MATERIALS AND METHODS Islets from deceased donors of both sexes and different ages were used for analysis. The expression status of senescence-related genes (glutaminase 1, interleukin 6, interleukin 8, cyclin-dependent kinase inhibitor 2A, cyclin-dependent kinase inhibitor 1A, and senescence-associated beta-galactosidase) and pancreatic function-related genes (glucagon and insulin) was examined by reverse transcription-quantitative polymerase chain reaction, and their relationships with age were investigated. RESULTS We obtained isolated human islets from 18 deceased multiorgan donors. There was no correlation between donor age and expression of any of the senescence-related genes. Regarding correlations between donor age and pancreatic function-related genes, age was positively correlated only with INS (r=0.49, p=0.03). INS expression was not correlated with that of GLS1 (r=0.23, p=0.34), IL6 (r=-0.06, p=0.79), or IL8 (r=-0.1, p=0.12), but positively related with p16 (r=0.89, p<0.0001), p21 (r=0.51, p=0.02), and SA-β-gal (r=0.52, p=0.02). CONCLUSION We showed the functional potential even of aged islets, which were originally thought to be functionally impaired. We were unable to identify any senescence-related genes expressed in islets from donors of different ages. Therefore, a new index is needed to evaluate not only actual chronological age but also organ- and cell-specific age.
Collapse
Affiliation(s)
- Hajime Imamura
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan;
| | - Tomohiko Adachi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan;
| | - Daisuke Miyamoto
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tatsuya Kin
- Clinical Islet Transplantation Program, University of Alberta, Edmonton, AB, Canada
| | - Mampei Yamashita
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hajime Matsushima
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takanobu Hara
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Akihiko Soyama
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Susumu Eguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
5
|
Zhang W, Wu L, Qu R, Liu T, Wang J, Tong Y, Bei W, Guo J, Hu X. Hesperidin activates the GLP-1R/cAMP-CREB/IRS2/PDX1 pathway to promote transdifferentiation of islet α cells into β cells Across the spectrum. Heliyon 2024; 10:e35424. [PMID: 39220963 PMCID: PMC11365324 DOI: 10.1016/j.heliyon.2024.e35424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/12/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Background and aims In all age, FoShou as a Chinese medicinal herb has been active in various kinds of Traditional Chinese medicine formula to treating diabetes. Hesperidin (HES), the main monomeric component of FoShou, has been extensively investigated for interventions with pathogenic mechanism of diabetes as well as subsequent treatment of associated complications. Islet β-cells have an essential effect on dynamically regulating blood sugar. Functional abnormalities in these cells and their death are strongly associated with the onset of diabetes. Therefore, induction of islet endocrine cell lineage re-editing for damaged βcell replenishment would be a promising therapeutic tool. Previously, it has been found that HES can protect islet β-cells in vivo, But, the regenerative function of HES in islet β cells and its role in promoting differential non-β cells transdifferentiation into β cells and cell fate rewriting associated mechanisms remain unclear.This work focused on investigating whether HES can induce islet α cells transdifferentiation into β cells for achieving damaged β cell regeneration and the causes and possible mechanisms involved in the process. Materials and methods In brief, 60 mg/kg/d streptozotocin (STZ) was administered intraperitoneally in each male C57bL/6J mouse raised by the high-sugar and high-fat diet (HFD) to create a diabetic mouse model with severe β-cell damage. After 28 consecutive days of HES treatment (160 mg/kg; 320 mg/kg; once daily, as appropriate). Tracing the dynamics of α as well as β cell transformation, together with β cells growth and apoptosis levels during treatment by cell lineage tracing. The self-enforcing transcriptional network on which the cell lineage is based is used as a clue to explore the underlying mechanisms. Guangdong Pharmaceutical University's Animal Experiment Ethics Committee (GDPulac2019180) approved all animal experiments. Results Localization by cell lineage we find that transdifferentiated newborn β-cells derived from α cells appeared in the islet endocrine cell mass of DM mice under HES'action. Compared to the model group, expressed by Tunel staining and CXCL10 levels the overall apoptosis rate of β-cells of the pancreas were reduced,the inflammatory infiltration feedback from HE staining were lower.Ki-67 positive cells showed enhanced β-cell proliferation. Decreased HbA1c and blood glucose contents, elevated C-Peptide and insulin contents which respond to ability of nascent beta cells. Also upregulated the mRNA levels of MafA, Ngn3, PDX-1, Pax4 and Arx. Moreover, increased the expression of TGR5/cAMP-CREB/GLP-1 in mouse intestinal tissues and GLP-1/GLP-1R and cAMP-CREB/IRS2/PDX-1 in pancreatic tissues. Conclusions HES directly affects β-cells, apart from being anti-apoptotic and reducing inflammatory infiltration. HES promotes GLP-1 release by intestinal L cells by activating the TGR5 receptor in DM mouse and regulating its response element CREB signaling. GLP-1 then uses the GLP-1/GLP-1R system to act on IRS2, IRS2 as a port to influence α precursor cells to express PDX-1, with the mobilization of Pax4 strong expression than Arx so that α cell lineage is finally reversed for achieving β cell endogenous proliferation.
Collapse
Affiliation(s)
- Wang Zhang
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Lele Wu
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ru Qu
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Tianfeng Liu
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jiliang Wang
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ying Tong
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Weijian Bei
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jiao Guo
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xuguang Hu
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
| |
Collapse
|
6
|
Zanfrini E, Bandral M, Jarc L, Ramirez-Torres MA, Pezzolla D, Kufrin V, Rodriguez-Aznar E, Avila AKM, Cohrs C, Speier S, Neumann K, Gavalas A. Generation and application of novel hES cell reporter lines for the differentiation and maturation of hPS cell-derived islet-like clusters. Sci Rep 2024; 14:19863. [PMID: 39191834 DOI: 10.1038/s41598-024-69645-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 08/07/2024] [Indexed: 08/29/2024] Open
Abstract
The significant advances in the differentiation of human pluripotent stem (hPS) cells into pancreatic endocrine cells, including functional β-cells, have been based on a detailed understanding of the underlying developmental mechanisms. However, the final differentiation steps, leading from endocrine progenitors to mono-hormonal and mature pancreatic endocrine cells, remain to be fully understood and this is reflected in the remaining shortcomings of the hPS cell-derived islet cells (SC-islet cells), which include a lack of β-cell maturation and variability among different cell lines. Additional signals and modifications of the final differentiation steps will have to be assessed in a combinatorial manner to address the remaining issues and appropriate reporter lines would be useful in this undertaking. Here we report the generation and functional validation of hPS cell reporter lines that can monitor the generation of INS+ and GCG+ cells and their resolution into mono-hormonal cells (INSeGFP, INSeGFP/GCGmCHERRY) as well as β-cell maturation (INSeGFP/MAFAmCHERRY) and function (INSGCaMP6). The reporter hPS cell lines maintained strong and widespread expression of pluripotency markers and differentiated efficiently into definitive endoderm and pancreatic progenitor (PP) cells. PP cells from all lines differentiated efficiently into islet cell clusters that robustly expressed the corresponding reporters and contained glucose-responsive, insulin-producing cells. To demonstrate the applicability of these hPS cell reporter lines in a high-content live imaging approach for the identification of optimal differentiation conditions, we adapted our differentiation procedure to generate SC-islet clusters in microwells. This allowed the live confocal imaging of multiple SC-islets for a single condition and, using this approach, we found that the use of the N21 supplement in the last stage of the differentiation increased the number of monohormonal β-cells without affecting the number of α-cells in the SC-islets. The hPS cell reporter lines and the high-content live imaging approach described here will enable the efficient assessment of multiple conditions for the optimal differentiation and maturation of SC-islets.
Collapse
Affiliation(s)
- Elisa Zanfrini
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Manuj Bandral
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Luka Jarc
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Maria Alejandra Ramirez-Torres
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Daniela Pezzolla
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Vida Kufrin
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Eva Rodriguez-Aznar
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Ana Karen Mojica Avila
- Institute of Physiology, Faculty of Medicine, TU Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Christian Cohrs
- Institute of Physiology, Faculty of Medicine, TU Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Stephan Speier
- Institute of Physiology, Faculty of Medicine, TU Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Katrin Neumann
- Stem Cell Engineering Facility (SCEF), CRTD, TU Dresden, Dresden, Germany
| | - Anthony Gavalas
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- German Centre for Diabetes Research (DZD), Munich-Neuherberg, Germany.
| |
Collapse
|
7
|
Wu Y, Qin K, Xu Y, Rajhans S, Vo T, Lopez KM, Liu J, Nipper MH, Deng J, Yin X, Ramjit LR, Ye Z, Luan Y, Arda HE, Wang P. Hippo pathway-mediated YAP1/TAZ inhibition is essential for proper pancreatic endocrine specification and differentiation. eLife 2024; 13:e84532. [PMID: 39051998 PMCID: PMC11272159 DOI: 10.7554/elife.84532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 06/15/2024] [Indexed: 07/27/2024] Open
Abstract
The Hippo pathway plays a central role in tissue development and homeostasis. However, the function of Hippo in pancreatic endocrine development remains obscure. Here, we generated novel conditional genetically engineered mouse models to examine the roles of Hippo pathway-mediated YAP1/TAZ inhibition in the development stages of endocrine specification and differentiation. While YAP1 protein was localized to the nuclei in bipotent progenitor cells, Neurogenin 3 expressing endocrine progenitors completely lost YAP1 expression. Using genetically engineered mouse models, we found that inactivation of YAP1 requires both an intact Hippo pathway and Neurogenin 3 protein. Gene deletion of Lats1 and 2 kinases (Lats1&2) in endocrine progenitor cells of developing mouse pancreas using Neurog3Cre blocked endocrine progenitor cell differentiation and specification, resulting in reduced islets size and a disorganized pancreas at birth. Loss of Lats1&2 in Neurogenin 3 expressing cells activated YAP1/TAZ transcriptional activity and recruited macrophages to the developing pancreas. These defects were rescued by deletion of Yap1/Wwtr1 genes, suggesting that tight regulation of YAP1/TAZ by Hippo signaling is crucial for pancreatic endocrine specification. In contrast, deletion of Lats1&2 using β-cell-specific Ins1CreER resulted in a phenotypically normal pancreas, indicating that Lats1&2 are indispensable for differentiation of endocrine progenitors but not for that of β-cells. Our results demonstrate that loss of YAP1/TAZ expression in the pancreatic endocrine compartment is not a passive consequence of endocrine specification. Rather, Hippo pathway-mediated inhibition of YAP1/TAZ in endocrine progenitors is a prerequisite for endocrine specification and differentiation.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
- Department of Obstetrics, The Second Xiangya Hospital, Central South UniversityChangshaChina
| | - Kunhua Qin
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
- Department of Molecular Medicine, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Yi Xu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Shreya Rajhans
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, NIHBethesdaUnited States
| | - Truong Vo
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, NIHBethesdaUnited States
| | - Kevin M Lopez
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Jun Liu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Michael H Nipper
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Janice Deng
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Xue Yin
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Logan R Ramjit
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Zhenqing Ye
- Department of Population Health Sciences, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Yu Luan
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - H Efsun Arda
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, NIHBethesdaUnited States
| | - Pei Wang
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| |
Collapse
|
8
|
Hill TG, Hill DJ. The Importance of Intra-Islet Communication in the Function and Plasticity of the Islets of Langerhans during Health and Diabetes. Int J Mol Sci 2024; 25:4070. [PMID: 38612880 PMCID: PMC11012451 DOI: 10.3390/ijms25074070] [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: 02/27/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Islets of Langerhans are anatomically dispersed within the pancreas and exhibit regulatory coordination between islets in response to nutritional and inflammatory stimuli. However, within individual islets, there is also multi-faceted coordination of function between individual beta-cells, and between beta-cells and other endocrine and vascular cell types. This is mediated partly through circulatory feedback of the major secreted hormones, insulin and glucagon, but also by autocrine and paracrine actions within the islet by a range of other secreted products, including somatostatin, urocortin 3, serotonin, glucagon-like peptide-1, acetylcholine, and ghrelin. Their availability can be modulated within the islet by pericyte-mediated regulation of microvascular blood flow. Within the islet, both endocrine progenitor cells and the ability of endocrine cells to trans-differentiate between phenotypes can alter endocrine cell mass to adapt to changed metabolic circumstances, regulated by the within-islet trophic environment. Optimal islet function is precariously balanced due to the high metabolic rate required by beta-cells to synthesize and secrete insulin, and they are susceptible to oxidative and endoplasmic reticular stress in the face of high metabolic demand. Resulting changes in paracrine dynamics within the islets can contribute to the emergence of Types 1, 2 and gestational diabetes.
Collapse
Affiliation(s)
- Thomas G. Hill
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - David J. Hill
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada;
- Departments of Medicine, Physiology and Pharmacology, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
9
|
Wei H, Ren H. Precision treatment of pancreatic ductal adenocarcinoma. Cancer Lett 2024; 585:216636. [PMID: 38278471 DOI: 10.1016/j.canlet.2024.216636] [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: 10/12/2023] [Revised: 12/08/2023] [Accepted: 01/07/2024] [Indexed: 01/28/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly heterogeneous tumor comprising pancreatic cancer cells, fibroblasts, immune cells, vascular epithelial cells, and other cells in the mesenchymal tissue. PDAC is difficult to treat because of the complexity of the tissue components; therefore, achieving therapeutic effects with a single therapeutic method or target is problematic. Recently, precision therapy has provided new directions and opportunities for treating PDAC using genetic information from an individual's disease to guide treatment. It selects and applies appropriate therapeutic methods for each patient, with an aim to minimize medical damage and costs, while maximizing patient benefits. Molecular targeted therapy is effective in most clinical studies; however, it has been ineffective in large-scale randomized controlled trials of PDAC, mainly because the enrolled populations were not stratified on a molecular basis. Molecular stratification allows the identification of the PDAC population being treated, optimizing therapeutic effect. However, a systematic review of precision therapies for patients with highly heterogeneous PDAC backgrounds has not been conducted. Here, we review the molecular background and current potential therapeutic targets related to PDAC and provide new directions for PDAC precision therapy.
Collapse
Affiliation(s)
- Hongyun Wei
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, China; Key Laboratory of Pancreatic Diseases, Center of Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, China.
| | - He Ren
- Key Laboratory of Pancreatic Diseases, Center of Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, China.
| |
Collapse
|
10
|
Lu P, Xu J, Shen X, Sun J, Liu M, Niu N, Wang Q, Xue J. Spatiotemporal role of SETD2-H3K36me3 in murine pancreatic organogenesis. Cell Rep 2024; 43:113703. [PMID: 38265933 DOI: 10.1016/j.celrep.2024.113703] [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: 01/04/2023] [Revised: 10/11/2023] [Accepted: 01/08/2024] [Indexed: 01/26/2024] Open
Abstract
Pancreas development is tightly controlled by multilayer mechanisms. Despite years of effort, large gaps remain in understanding how histone modifications coordinate pancreas development. SETD2, a predominant histone methyltransferase of H3K36me3, plays a key role in embryonic stem cell differentiation, whose role in organogenesis remains elusive. Here, by combination of cleavage under targets and tagmentation (CUT&Tag), assay for transposase-accessible chromatin using sequencing (ATAC-seq), and bulk RNA sequencing, we show a dramatic increase in the H3K36me3 level from the secondary transition phase and decipher the related transcriptional alteration. Using single-cell RNA sequencing, we define that pancreatic deletion of Setd2 results in abnormalities in both exocrine and endocrine lineages: hyperproliferative tip progenitor cells lead to abnormal differentiation; Ngn3+ endocrine progenitors decline due to the downregulation of Nkx2.2, leading to insufficient endocrine development. Thus, these data identify SETD2 as a crucial player in embryonic pancreas development, providing a clue to understanding the dysregulation of histone modifications in pancreatic disorders.
Collapse
Affiliation(s)
- Ping Lu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Junyi Xu
- Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuqing Shen
- Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiajun Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingzhu Liu
- Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ningning Niu
- Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qidi Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jing Xue
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
11
|
Jarc L, Bandral M, Zanfrini E, Lesche M, Kufrin V, Sendra R, Pezzolla D, Giannios I, Khattak S, Neumann K, Ludwig B, Gavalas A. Regulation of multiple signaling pathways promotes the consistent expansion of human pancreatic progenitors in defined conditions. eLife 2024; 12:RP89962. [PMID: 38180318 PMCID: PMC10945307 DOI: 10.7554/elife.89962] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024] Open
Abstract
The unlimited expansion of human progenitor cells in vitro could unlock many prospects for regenerative medicine. However, it remains an important challenge as it requires the decoupling of the mechanisms supporting progenitor self-renewal and expansion from those mechanisms promoting their differentiation. This study focuses on the expansion of human pluripotent stem (hPS) cell-derived pancreatic progenitors (PP) to advance novel therapies for diabetes. We obtained mechanistic insights into PP expansion requirements and identified conditions for the robust and unlimited expansion of hPS cell-derived PP cells under GMP-compliant conditions through a hypothesis-driven iterative approach. We show that the combined stimulation of specific mitogenic pathways, suppression of retinoic acid signaling, and inhibition of selected branches of the TGFβ and Wnt signaling pathways are necessary for the effective decoupling of PP proliferation from differentiation. This enabled the reproducible, 2000-fold, over 10 passages and 40-45 d, expansion of PDX1+/SOX9+/NKX6-1+ PP cells. Transcriptome analyses confirmed the stabilization of PP identity and the effective suppression of differentiation. Using these conditions, PDX1+/SOX9+/NKX6-1+ PP cells, derived from different, both XY and XX, hPS cell lines, were enriched to nearly 90% homogeneity and expanded with very similar kinetics and efficiency. Furthermore, non-expanded and expanded PP cells, from different hPS cell lines, were differentiated in microwells into homogeneous islet-like clusters (SC-islets) with very similar efficiency. These clusters contained abundant β-cells of comparable functionality as assessed by glucose-stimulated insulin secretion assays. These findings established the signaling requirements to decouple PP proliferation from differentiation and allowed the consistent expansion of hPS cell-derived PP cells. They will enable the establishment of large banks of GMP-produced PP cells derived from diverse hPS cell lines. This approach will streamline SC-islet production for further development of the differentiation process, diabetes research, personalized medicine, and cell therapies.
Collapse
Affiliation(s)
- Luka Jarc
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental HealthNeuherbergGermany
- German Centre for Diabetes Research (DZD)MunichGermany
| | - Manuj Bandral
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental HealthNeuherbergGermany
- German Centre for Diabetes Research (DZD)MunichGermany
| | - Elisa Zanfrini
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental HealthNeuherbergGermany
- German Centre for Diabetes Research (DZD)MunichGermany
| | - Mathias Lesche
- Dresden Concept Genome Centre (DcGC), TU DresdenDresdenGermany
- Center for Molecular and Cellular Bioengineering (CMCB) Technology Platform, TU DresdenDresdenGermany
| | - Vida Kufrin
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental HealthNeuherbergGermany
| | - Raquel Sendra
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental HealthNeuherbergGermany
| | - Daniela Pezzolla
- German Centre for Diabetes Research (DZD)MunichGermany
- Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, TU DresdenDresdenGermany
| | - Ioannis Giannios
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental HealthNeuherbergGermany
- German Centre for Diabetes Research (DZD)MunichGermany
| | - Shahryar Khattak
- Stem Cell Engineering Facility, (SCEF), CRTD, Faculty of Medicine, TU DresdenDresdenGermany
| | - Katrin Neumann
- Stem Cell Engineering Facility, (SCEF), CRTD, Faculty of Medicine, TU DresdenDresdenGermany
| | - Barbara Ludwig
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental HealthNeuherbergGermany
- German Centre for Diabetes Research (DZD)MunichGermany
- Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, TU DresdenDresdenGermany
- Department of Medicine III, University Hospital Carl Gustav Carus and Faculty of Medicine, TU DresdenDresdenGermany
| | - Anthony Gavalas
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental HealthNeuherbergGermany
- German Centre for Diabetes Research (DZD)MunichGermany
| |
Collapse
|
12
|
Sepyani S, Momenzadeh S, Safabakhsh S, Nedaeinia R, Salehi R. Therapeutic approaches for Type 1 Diabetes: Promising cell-based approaches to achieve ultimate success. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:23-33. [PMID: 37977308 DOI: 10.1016/j.slasd.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/12/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Type 1 Diabetes mellitus (T1DM) is a chronic metabolic disorder characterized by pancreatic β-cells destruction. Despite substantial advances in T1DM treatment, lifelong exogenous insulin administration is the mainstay of treatments, and constant control of glucose levels is still a challenge. Endogenous insulin production by replacing insulin-producing cells is an alternative, but the lack of suitable donors is accounted as one of the main obstacles to its widespread application. The research and trials overview demonstrates that endogenous production of insulin has started to go beyond the deceased-derived to stem cells-derived insulin-producing cells. Several protocols have been developed over the past couple of years for generating insulin-producing cells (IPCs) from various stem cell types and reprogramming fully differentiated cells. A straightforward and quick method for achieving this goal is to investigate and apply the β-cell specific transcription factors as a direct strategy for IPCs generation. In this review, we emphasize the significance of transcription factors in IPCs development from different non-beta cell sources, and pertinent research underlies the marked progress in the methods for generating insulin-producing cells and application for Type 1 Diabetes treatment.
Collapse
Affiliation(s)
- Sahar Sepyani
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sedigheh Momenzadeh
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saied Safabakhsh
- Micronesian Institute for Disease Prevention and Research, 736 Route 4, Suite 103, Sinajana, GU 96910, United States
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rasoul Salehi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
13
|
Cota P, Caliskan ÖS, Bastidas-Ponce A, Jing C, Jaki J, Saber L, Czarnecki O, Taskin D, Blöchinger AK, Kurth T, Sterr M, Burtscher I, Krahmer N, Lickert H, Bakhti M. Insulin regulates human pancreatic endocrine cell differentiation in vitro. Mol Metab 2024; 79:101853. [PMID: 38103636 PMCID: PMC10765254 DOI: 10.1016/j.molmet.2023.101853] [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] [Received: 09/14/2023] [Revised: 11/21/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
OBJECTIVE The consequences of mutations in genes associated with monogenic forms of diabetes on human pancreas development cannot be studied in a time-resolved fashion in vivo. More specifically, if recessive mutations in the insulin gene influence human pancreatic endocrine lineage formation is still an unresolved question. METHODS To model the extremely reduced insulin levels in patients with recessive insulin gene mutations, we generated a novel knock-in H2B-Cherry reporter human induced pluripotent stem cell (iPSC) line expressing no insulin upon differentiation to stem cell-derived (SC-) β cells in vitro. Differentiation of iPSCs into the pancreatic and endocrine lineage, combined with immunostaining, Western blotting and proteomics analysis phenotypically characterized the insulin gene deficiency in SC-islets. Furthermore, we leveraged FACS analysis and confocal microscopy to explore the impact of insulin shortage on human endocrine cell induction, composition, differentiation and proliferation. RESULTS Interestingly, insulin-deficient SC-islets exhibited low insulin receptor (IR) signaling when stimulated with glucose but displayed increased IR sensitivity upon treatment with exogenous insulin. Furthermore, insulin shortage did not alter neurogenin-3 (NGN3)-mediated endocrine lineage induction. Nevertheless, lack of insulin skewed the SC-islet cell composition with an increased number in SC-β cell formation at the expense of SC-α cells. Finally, insulin deficiency reduced the rate of SC-β cell proliferation but had no impact on the expansion of SC-α cells. CONCLUSIONS Using iPSC disease modelling, we provide first evidence of insulin function in human pancreatic endocrine lineage formation. These findings help to better understand the phenotypic impact of recessive insulin gene mutations during pancreas development and shed light on insulin gene function beside its physiological role in blood glucose regulation.
Collapse
Affiliation(s)
- Perla Cota
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Özüm Sehnaz Caliskan
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute of Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Changying Jing
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Munich medical research school (MMRS), Ludwig Maximilian University (LMU), Munich, Germany
| | - Jessica Jaki
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Lama Saber
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Oliver Czarnecki
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Damla Taskin
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
| | - Anna Karolina Blöchinger
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Thomas Kurth
- Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform Core Facility Electron Microscopy and Histology, Technische Universität Dresden, Dresden, Germany
| | - Michael Sterr
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Natalie Krahmer
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute of Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany.
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
14
|
Yu V, Yong F, Marta A, Khadayate S, Osakwe A, Bhattacharya S, Varghese SS, Chabosseau P, Tabibi SM, Chen K, Georgiadou E, Parveen N, Suleiman M, Stamoulis Z, Marselli L, De Luca C, Tesi M, Ostinelli G, Delgadillo-Silva L, Wu X, Hatanaka Y, Montoya A, Elliott J, Patel B, Demchenko N, Whilding C, Hajkova P, Shliaha P, Kramer H, Ali Y, Marchetti P, Sladek R, Dhawan S, Withers DJ, Rutter GA, Millership SJ. Differential CpG methylation at Nnat in the early establishment of beta cell heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.04.527050. [PMID: 38076935 PMCID: PMC10705251 DOI: 10.1101/2023.02.04.527050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Aims/hypothesis Beta cells within the pancreatic islet represent a heterogenous population wherein individual sub-groups of cells make distinct contributions to the overall control of insulin secretion. These include a subpopulation of highly-connected 'hub' cells, important for the propagation of intercellular Ca2+ waves. Functional subpopulations have also been demonstrated in human beta cells, with an altered subtype distribution apparent in type 2 diabetes. At present, the molecular mechanisms through which beta cell hierarchy is established are poorly understood. Changes at the level of the epigenome provide one such possibility which we explore here by focussing on the imprinted gene neuronatin (Nnat), which is required for normal insulin synthesis and secretion. Methods Single cell RNA-seq datasets were examined using Seurat 4.0 and ClusterProfiler running under R. Transgenic mice expressing eGFP under the control of the Nnat enhancer/promoter regions were generated for fluorescence-activated cell (FAC) sorting of beta cells and downstream analysis of CpG methylation by bisulphite and RNA sequencing, respectively. Animals deleted for the de novo methyltransferase, DNMT3A from the pancreatic progenitor stage were used to explore control of promoter methylation. Proteomics was performed using affinity purification mass spectrometry and Ca2+ dynamics explored by rapid confocal imaging of Cal-520 and Cal-590. Insulin secretion was measured using Homogeneous Time Resolved Fluorescence Imaging. Results Nnat mRNA was differentially expressed in a discrete beta cell population in a developmental stage- and DNA methylation (DNMT3A)-dependent manner. Thus, pseudo-time analysis of embryonic data sets demonstrated the early establishment of Nnat-positive and negative subpopulations during embryogenesis. NNAT expression is also restricted to a subset of beta cells across the human islet that is maintained throughout adult life. NNAT+ beta cells also displayed a discrete transcriptome at adult stages, representing a sub-population specialised for insulin production, reminiscent of recently-described "βHI" cells and were diminished in db/db mice. 'Hub' cells were less abundant in the NNAT+ population, consistent with epigenetic control of this functional specialization. Conclusions/interpretation These findings demonstrate that differential DNA methylation at Nnat represents a novel means through which beta cell heterogeneity is established during development. We therefore hypothesise that changes in methylation at this locus may thus contribute to a loss of beta cell hierarchy and connectivity, potentially contributing to defective insulin secretion in some forms of diabetes.
Collapse
Affiliation(s)
- Vanessa Yu
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Fiona Yong
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, 637553, Singapore
| | - Angellica Marta
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Sanjay Khadayate
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Adrien Osakwe
- Departments of Medicine, Human Genetics and Quantitative Life Sciences, McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
| | - Supriyo Bhattacharya
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Sneha S. Varghese
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Pauline Chabosseau
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Sayed M. Tabibi
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Keran Chen
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Eleni Georgiadou
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Nazia Parveen
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy
| | - Zoe Stamoulis
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy
| | - Carmela De Luca
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy
| | - Marta Tesi
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy
| | - Giada Ostinelli
- CHUM Research Center and Faculty of Medicine, University of Montréal, 900 Rue St Denis, Montréal, H2X OA9, QC, Canada
| | - Luis Delgadillo-Silva
- CHUM Research Center and Faculty of Medicine, University of Montréal, 900 Rue St Denis, Montréal, H2X OA9, QC, Canada
| | - Xiwei Wu
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Yuki Hatanaka
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Alex Montoya
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - James Elliott
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Bhavik Patel
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Nikita Demchenko
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Chad Whilding
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Petra Hajkova
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Pavel Shliaha
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Holger Kramer
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Yusuf Ali
- Nutrition, Metabolism and Health Programme & Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, 308232
- Singapore Eye Research Institute (SERI), Singapore General Hospital, Singapore, 168751
- Clinical Research Unit, Khoo Teck Puat Hospital, National Healthcare Group, Singapore, 768828
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, and AOUP Cisanello University Hospital, University of Pisa, Pisa 56126, Italy
| | - Robert Sladek
- Departments of Medicine, Human Genetics and Quantitative Life Sciences, McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
| | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Dominic J. Withers
- MRC Laboratory of Medical Sciences, Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Guy A. Rutter
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, 637553, Singapore
- CHUM Research Center and Faculty of Medicine, University of Montréal, 900 Rue St Denis, Montréal, H2X OA9, QC, Canada
| | - Steven J. Millership
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
15
|
Tao Z, Yang D, Ni R. Tmed10 deficiency results in impaired exocrine pancreatic differentiation in zebrafish larvae. Dev Biol 2023; 503:43-52. [PMID: 37597605 DOI: 10.1016/j.ydbio.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 08/01/2023] [Accepted: 08/15/2023] [Indexed: 08/21/2023]
Abstract
Transmembrane p24 trafficking protein 10 (TMED10) is a conserved vesicle trafficking protein. It is dysregulated in Alzheimer disease and plays a pivotal role in the pathogenesis of Alzheimer disease. In addition to the brain, TMED10 is highly expressed in the exocrine pancreas; however, its biological functions and underlying mechanisms remain largely unknown. We studied reduced Tmed10 in zebrafish embryos by morpholino oligonucleotide knockdown and CRISPR-Cas9 mutagenesis. Tmed10-deficient embryos showed extensive loss of acinar mass and impaired acinar differentiation. TMED10 has been reported to have an inhibitory effect on γ-secretase. As one of the substrates of γ-secretase, membrane-bound β-catenin was significantly reduced in Tmed10-deficient embryos. Increased γ-secretase activity in wild-type embryos resulted in a phenotype similar to that of tmed10 mutants. And the mutant phenotype could be rescued by treatment with the γ-secretase inhibitor, N-[N-(3, 5-difluorophenacetyl)-l-alanyl]-s-phenylglycinet-butyl ester (DAPT). In addition, the reduced membrane-bound β-catenin was accompanied with up-regulated β-catenin target genes in Tmed10-deficient embryos. Overexpression of β-catenin signaling inhibitor Dickkopf-1 (DKK-1) could rescue the exocrine pancreas defects. Taken together, our study reveals that Tmed10 regulates exocrine pancreatic differentiation through γ-secretase. Reduced membrane-bound β-catenin, accompanied with hyperactivation of β-catenin signaling, is an important cause of exocrine pancreas defects in Tmed10-deficient embryos. Our study reaffirms the importance of appropriate β-catenin signaling in exocrine pancreas development. These findings may provide a theoretical basis for the development of treatment strategies for TMED10-related diseases.
Collapse
Affiliation(s)
- Zewen Tao
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, 400715, Chongqing, China
| | - Di Yang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, 400715, Chongqing, China
| | - Rui Ni
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, 400715, Chongqing, China.
| |
Collapse
|
16
|
Cota P, Saber L, Taskin D, Jing C, Bastidas-Ponce A, Vanheusden M, Shahryari A, Sterr M, Burtscher I, Bakhti M, Lickert H. NEUROD2 function is dispensable for human pancreatic β cell specification. Front Endocrinol (Lausanne) 2023; 14:1286590. [PMID: 37955006 PMCID: PMC10634430 DOI: 10.3389/fendo.2023.1286590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Introduction The molecular programs regulating human pancreatic endocrine cell induction and fate allocation are not well deciphered. Here, we investigated the spatiotemporal expression pattern and the function of the neurogenic differentiation factor 2 (NEUROD2) during human endocrinogenesis. Methods Using Crispr-Cas9 gene editing, we generated a reporter knock-in transcription factor (TF) knock-out human inducible pluripotent stem cell (iPSC) line in which the open reading frame of both NEUROD2 alleles are replaced by a nuclear histone 2B-Venus reporter (NEUROD2nVenus/nVenus). Results We identified a transient expression of NEUROD2 mRNA and its nuclear Venus reporter activity at the stage of human endocrine progenitor formation in an iPSC differentiation model. This expression profile is similar to what was previously reported in mice, uncovering an evolutionarily conserved gene expression pattern of NEUROD2 during endocrinogenesis. In vitro differentiation of the generated homozygous NEUROD2nVenus/nVenus iPSC line towards human endocrine lineages uncovered no significant impact upon the loss of NEUROD2 on endocrine cell induction. Moreover, analysis of endocrine cell specification revealed no striking changes in the generation of insulin-producing b cells and glucagon-secreting a cells upon lack of NEUROD2. Discussion Overall, our results suggest that NEUROD2 is expendable for human b cell formation in vitro.
Collapse
Affiliation(s)
- Perla Cota
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Lama Saber
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Damla Taskin
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
| | - Changying Jing
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Munich Medical Research School (MMRS), Ludwig Maximilian University (LMU), Munich, Germany
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Matthew Vanheusden
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
| | - Alireza Shahryari
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
| | - Michael Sterr
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- School of Medicine, Technical University of Munich (TUM), Munich, Germany
| |
Collapse
|
17
|
Abstract
Nutrient intake is obligatory for animal growth and development, but nutrients alone are not sufficient. Indeed, insulin and homologous hormones are required for normal growth even in the presence of nutrients. These hormones communicate nutrient status between organs, allowing animals to coordinate growth and metabolism with nutrient supply. Insulin and related hormones, such as insulin-like growth factors and insulin-like peptides, play important roles in development and metabolism, with defects in insulin production and signaling leading to hyperglycemia and diabetes. Here, we describe the insulin hormone family and the signal transduction pathways activated by these hormones. We highlight the roles of insulin signaling in coordinating maternal and fetal metabolism and growth during pregnancy, and we describe how secretion of insulin is regulated at different life stages. Additionally, we discuss the roles of insulin signaling in cell growth, stem cell proliferation and cell differentiation. We provide examples of the role of insulin in development across multiple model organisms: Caenorhabditis elegans, Drosophila, zebrafish, mouse and human.
Collapse
Affiliation(s)
- Miyuki Suzawa
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Michelle L. Bland
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
18
|
Bohuslavova R, Fabriciova V, Smolik O, Lebrón-Mora L, Abaffy P, Benesova S, Zucha D, Valihrach L, Berkova Z, Saudek F, Pavlinkova G. NEUROD1 reinforces endocrine cell fate acquisition in pancreatic development. Nat Commun 2023; 14:5554. [PMID: 37689751 PMCID: PMC10492842 DOI: 10.1038/s41467-023-41306-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023] Open
Abstract
NEUROD1 is a transcription factor that helps maintain a mature phenotype of pancreatic β cells. Disruption of Neurod1 during pancreatic development causes severe neonatal diabetes; however, the exact role of NEUROD1 in the differentiation programs of endocrine cells is unknown. Here, we report a crucial role of the NEUROD1 regulatory network in endocrine lineage commitment and differentiation. Mechanistically, transcriptome and chromatin landscape analyses demonstrate that Neurod1 inactivation triggers a downregulation of endocrine differentiation transcription factors and upregulation of non-endocrine genes within the Neurod1-deficient endocrine cell population, disturbing endocrine identity acquisition. Neurod1 deficiency altered the H3K27me3 histone modification pattern in promoter regions of differentially expressed genes, which resulted in gene regulatory network changes in the differentiation pathway of endocrine cells, compromising endocrine cell potential, differentiation, and functional properties.
Collapse
Affiliation(s)
- Romana Bohuslavova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Valeria Fabriciova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Ondrej Smolik
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Laura Lebrón-Mora
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Sarka Benesova
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Daniel Zucha
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Zuzana Berkova
- Diabetes Centre, Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, 14021, Prague, Czechia
| | - Frantisek Saudek
- Diabetes Centre, Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, 14021, Prague, Czechia
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia.
| |
Collapse
|
19
|
Braam MJS, Zhao J, Liang S, Ida S, Kloostra NK, Iworima DG, Tang M, Baker RK, Quiskamp N, Piret JM, Kieffer TJ. Protocol development to further differentiate and transition stem cell-derived pancreatic progenitors from a monolayer into endocrine cells in suspension culture. Sci Rep 2023; 13:8877. [PMID: 37264038 DOI: 10.1038/s41598-023-35716-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/23/2023] [Indexed: 06/03/2023] Open
Abstract
The generation of functional β-cells from human pluripotent stem cells (hPSCs) for cell replacement therapy and disease modeling of diabetes is being investigated by many groups. We have developed a protocol to harvest and aggregate hPSC-derived pancreatic progenitors generated using a commercially available kit into near uniform spheroids and to further differentiate the cells toward an endocrine cell fate in suspension culture. Using a static suspension culture platform, we could generate a high percentage of insulin-expressing, glucose-responsive cells. We identified FGF7 as a soluble factor promoting aggregate survival with no inhibitory effect on endocrine gene expression. Notch inhibition of pancreatic progenitor cells during aggregation improved endocrine cell induction in vitro and improved graft function following implantation and further differentiation in mice. Thus we provide an approach to promote endocrine formation from kit-derived pancreatic progenitors, either through extended culture or post implant.
Collapse
Affiliation(s)
- Mitchell J S Braam
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Jia Zhao
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Shenghui Liang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Shogo Ida
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Nick K Kloostra
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Diepiriye G Iworima
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Mei Tang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Robert K Baker
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | | | - James M Piret
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Timothy J Kieffer
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
20
|
Gopalan V, Hannenhalli S. Towards a Synthesis of the Non-Genetic and Genetic Views of Cancer in Understanding Pancreatic Ductal Adenocarcinoma Initiation and Prevention. Cancers (Basel) 2023; 15:cancers15072159. [PMID: 37046820 PMCID: PMC10093726 DOI: 10.3390/cancers15072159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
While much of the research in oncogenesis and cancer therapy has focused on mutations in key cancer driver genes, more recent work suggests a complementary non-genetic paradigm. This paradigm focuses on how transcriptional and phenotypic heterogeneity, even in clonally derived cells, can create sub-populations associated with oncogenesis, metastasis, and therapy resistance. We discuss this complementary paradigm in the context of pancreatic ductal adenocarcinoma. A better understanding of cellular transcriptional heterogeneity and its association with oncogenesis can lead to more effective therapies that prevent tumor initiation and slow progression.
Collapse
Affiliation(s)
- Vishaka Gopalan
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| |
Collapse
|
21
|
Đorđević M, Stepper P, Feuerstein-Akgoz C, Gerhauser C, Paunović V, Tolić A, Rajić J, Dinić S, Uskoković A, Grdović N, Mihailović M, Jurkowska RZ, Jurkowski TP, Jovanović JA, Vidaković M. EpiCRISPR targeted methylation of Arx gene initiates transient switch of mouse pancreatic alpha to insulin-producing cells. Front Endocrinol (Lausanne) 2023; 14:1134478. [PMID: 37008919 PMCID: PMC10063207 DOI: 10.3389/fendo.2023.1134478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction Beta cell dysfunction by loss of beta cell identity, dedifferentiation, and the presence of polyhormonal cells are main characteristics of diabetes. The straightforward strategy for curing diabetes implies reestablishment of pancreatic beta cell function by beta cell replacement therapy. Aristaless-related homeobox (Arx) gene encodes protein which plays an important role in the development of pancreatic alpha cells and is a main target for changing alpha cell identity. Results In this study we used CRISPR/dCas9-based epigenetic tools for targeted hypermethylation of Arx gene promoter and its subsequent suppression in mouse pancreatic αTC1-6 cell line. Bisulfite sequencing and methylation profiling revealed that the dCas9-Dnmt3a3L-KRAB single chain fusion constructs (EpiCRISPR) was the most efficient. Epigenetic silencing of Arx expression was accompanied by an increase in transcription of the insulin gene (Ins2) mRNA on 5th and 7th post-transfection day, quantified by both RT-qPCR and RNA-seq. Insulin production and secretion was determined by immunocytochemistry and ELISA assay, respectively. Eventually, we were able to induce switch of approximately 1% of transiently transfected cells which were able to produce 35% more insulin than Mock transfected alpha cells. Conclusion In conclusion, we successfully triggered a direct, transient switch of pancreatic alpha to insulin-producing cells opening a future research on promising therapeutic avenue for diabetes management.
Collapse
Affiliation(s)
- Marija Đorđević
- Department of Molecular Biology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Peter Stepper
- Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, Stuttgart, Germany
| | - Clarissa Feuerstein-Akgoz
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Clarissa Gerhauser
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Verica Paunović
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Anja Tolić
- Department of Molecular Biology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jovana Rajić
- Department of Molecular Biology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Svetlana Dinić
- Department of Molecular Biology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Uskoković
- Department of Molecular Biology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Nevena Grdović
- Department of Molecular Biology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Mirjana Mihailović
- Department of Molecular Biology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | | | | | - Jelena Arambašić Jovanović
- Department of Molecular Biology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Melita Vidaković
- Department of Molecular Biology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
22
|
Leavens KF, Alvarez-Dominguez JR, Vo LT, Russ HA, Parent AV. Stem cell-based multi-tissue platforms to model human autoimmune diabetes. Mol Metab 2022; 66:101610. [PMID: 36209784 PMCID: PMC9587366 DOI: 10.1016/j.molmet.2022.101610] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/20/2022] [Accepted: 10/04/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is an autoimmune disease in which pancreatic insulin-producing β cells are specifically destroyed by the immune system. Understanding the initiation and progression of human T1D has been hampered by the lack of appropriate models that can reproduce the complexity and heterogeneity of the disease. The development of platforms combining multiple human pluripotent stem cell (hPSC) derived tissues to model distinct aspects of T1D has the potential to provide critical novel insights into the etiology and pathogenesis of the human disease. SCOPE OF REVIEW In this review, we summarize the state of hPSC differentiation approaches to generate cell types and tissues relevant to T1D, with a particular focus on pancreatic islet cells, T cells, and thymic epithelium. We present current applications as well as limitations of using these hPSC-derived cells for disease modeling and discuss efforts to optimize platforms combining multiple cell types to model human T1D. Finally, we outline remaining challenges and emphasize future improvements needed to accelerate progress in this emerging field of research. MAJOR CONCLUSIONS Recent advances in reprogramming approaches to create patient-specific induced pluripotent stem cell lines (iPSCs), genome engineering technologies to efficiently modify DNA of hPSCs, and protocols to direct their differentiation into mature cell types have empowered the use of stem cell derivatives to accurately model human disease. While challenges remain before complex interactions occurring in human T1D can be modeled with these derivatives, experiments combining hPSC-derived β cells and immune cells are already providing exciting insight into how these cells interact in the context of T1D, supporting the viability of this approach.
Collapse
Affiliation(s)
- Karla F Leavens
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania and Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Juan R Alvarez-Dominguez
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Linda T Vo
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Holger A Russ
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Audrey V Parent
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
23
|
Paganos P, Ronchi P, Carl J, Mizzon G, Martinez P, Benvenuto G, Arnone MI. Integrating single cell transcriptomics and volume electron microscopy confirms the presence of pancreatic acinar-like cells in sea urchins. Front Cell Dev Biol 2022; 10:991664. [PMID: 36060803 PMCID: PMC9437490 DOI: 10.3389/fcell.2022.991664] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 07/21/2022] [Indexed: 01/11/2023] Open
Abstract
The identity and function of a given cell type relies on the differential expression of gene batteries that promote diverse phenotypes and functional specificities. Therefore, the identification of the molecular and morphological fingerprints of cell types across taxa is essential for untangling their evolution. Here we use a multidisciplinary approach to identify the molecular and morphological features of an exocrine, pancreas-like cell type harbored within the sea urchin larval gut. Using single cell transcriptomics, we identify various cell populations with a pancreatic-like molecular fingerprint that are enriched within the S. purpuratus larva digestive tract. Among these, in the region where they reside, the midgut/stomach domain, we find that populations of exocrine pancreas-like cells have a unique regulatory wiring distinct from the rest the of the cell types of the same region. Furthermore, Serial Block-face scanning Electron Microscopy (SBEM) of the exocrine cells shows that this reported molecular diversity is associated to distinct morphological features that reflect the physiological and functional properties of this cell type. Therefore, we propose that these sea urchin exocrine cells are homologous to the well-known mammalian pancreatic acinar cells and thus we trace the origin of this particular cell type to the time of deuterostome diversification. Overall, our approach allows a thorough characterization of a complex cell type and shows how both the transcriptomic and morphological information contribute to disentangling the evolution of cell types and organs such as the pancreatic cells and pancreas.
Collapse
Affiliation(s)
| | - Paolo Ronchi
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Jil Carl
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Giulia Mizzon
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Pedro Martinez
- Institut Català de Recerca i Estudis Avancats (ICREA), Barcelona, Spain,Genetics Department, University of Barcelona, Barcelona, Spain
| | | | - Maria Ina Arnone
- Stazione Zoologica Anton Dohrn (SZN), Naples, Italy,*Correspondence: Maria Ina Arnone,
| |
Collapse
|
24
|
N 6-methyladenosine modification-mediated mRNA metabolism is essential for human pancreatic lineage specification and islet organogenesis. Nat Commun 2022; 13:4148. [PMID: 35851388 PMCID: PMC9293889 DOI: 10.1038/s41467-022-31698-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 06/27/2022] [Indexed: 11/30/2022] Open
Abstract
Pancreatic differentiation from human pluripotent stem cells (hPSCs) provides promising avenues for investigating development and treating diseases. N6-methyladenosine (m6A) is the most prevalent internal messenger RNA (mRNA) modification and plays pivotal roles in regulation of mRNA metabolism, while its functions remain elusive. Here, we profile the dynamic landscapes of m6A transcriptome-wide during pancreatic differentiation. Next, we generate knockout hPSC lines of the major m6A demethylase ALKBH5, and find that ALKBH5 plays significant regulatory roles in pancreatic organogenesis. Mechanistic studies reveal that ALKBH5 deficiency reduces the mRNA stability of key pancreatic transcription factors in an m6A and YTHDF2-dependent manner. We further identify that ALKBH5 cofactor α-ketoglutarate can be applied to enhance differentiation. Collectively, our findings identify ALKBH5 as an essential regulator of pancreatic differentiation and highlight that m6A modification-mediated mRNA metabolism presents an important layer of regulation during cell-fate specification and holds great potentials for translational applications. Ma et al. profile the dynamic landscape of m6A during pancreatic differentiation, and identify ALKBH5 as an essential m6A regulator supporting pancreatic differentiation, indicating a role for m6A-mediated mRNA metabolism in cell-fate specification.
Collapse
|
25
|
Duvall E, Benitez CM, Tellez K, Enge M, Pauerstein PT, Li L, Baek S, Quake SR, Smith JP, Sheffield NC, Kim SK, Arda HE. Single-cell transcriptome and accessible chromatin dynamics during endocrine pancreas development. Proc Natl Acad Sci U S A 2022; 119:e2201267119. [PMID: 35733248 PMCID: PMC9245718 DOI: 10.1073/pnas.2201267119] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 05/10/2022] [Indexed: 12/24/2022] Open
Abstract
Delineating gene regulatory networks that orchestrate cell-type specification is a continuing challenge for developmental biologists. Single-cell analyses offer opportunities to address these challenges and accelerate discovery of rare cell lineage relationships and mechanisms underlying hierarchical lineage decisions. Here, we describe the molecular analysis of mouse pancreatic endocrine cell differentiation using single-cell transcriptomics, chromatin accessibility assays coupled to genetic labeling, and cytometry-based cell purification. We uncover transcription factor networks that delineate β-, α-, and δ-cell lineages. Through genomic footprint analysis, we identify transcription factor-regulatory DNA interactions governing pancreatic cell development at unprecedented resolution. Our analysis suggests that the transcription factor Neurog3 may act as a pioneer transcription factor to specify the pancreatic endocrine lineage. These findings could improve protocols to generate replacement endocrine cells from renewable sources, like stem cells, for diabetes therapy.
Collapse
Affiliation(s)
- Eliza Duvall
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Cecil M. Benitez
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Krissie Tellez
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Martin Enge
- Department of Bioengineering and Applied Physics, Stanford University, Stanford, CA 94305
| | - Philip T. Pauerstein
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Lingyu Li
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Songjoon Baek
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Stephen R. Quake
- Department of Bioengineering and Applied Physics, Stanford University, Stanford, CA 94305
- Chan Zuckerberg Biohub, San Francisco, CA 94158
| | - Jason P. Smith
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908
| | - Nathan C. Sheffield
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908
| | - Seung K. Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305
| | - H. Efsun Arda
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892
| |
Collapse
|
26
|
Dang Le Q, Rodprasert W, Kuncorojakti S, Pavasant P, Osathanon T, Sawangmake C. In vitro generation of transplantable insulin-producing cells from canine adipose-derived mesenchymal stem cells. Sci Rep 2022; 12:9127. [PMID: 35650303 PMCID: PMC9160001 DOI: 10.1038/s41598-022-13114-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/20/2022] [Indexed: 11/27/2022] Open
Abstract
Canine mesenchymal stem cells (cMSCs) have potential applications for regenerative therapy, including the generation of insulin-producing cells (IPCs) for studying and treating diabetes. In this study, we established a useful protocol for generating IPCs from canine adipose mesenchymal stem cells (cAD-MSCs). Subsequently, in vitro preservation of pluronic F127-coated alginate (ALGPA)-encapsulated cAD-MSC-derived IPCs was performed to verify ready-to-use IPCs. IPCs were induced from cAD-MSCs with the modulated three-stepwise protocol. The first step of definitive endoderm (DE) induction showed that the cooperation of Chir99021 and Activin A created the effective production of Sox17-expressed DE cells. The second step for pancreatic endocrine (PE) progenitor induction from DE indicated that the treatment with taurine, retinoic acid, FGF2, EGF, TGFβ inhibitor, dorsomorphin, nicotinamide, and DAPT showed the significant upregulation of the pancreatic endocrine precursor markers Pdx1 and Ngn3. The last step of IPC production, the combination of taurine, nicotinamide, Glp-1, forskolin, PI3K inhibitor, and TGFβ inhibitor, yielded efficiently functional IPCs from PE precursors. Afterward, the maintenance of ALGPA-encapsulated cAD-MSC-derived IPCs with VSCBIC-1, a specialized medium, enhanced IPC properties. Conclusion, the modulated three-stepwise protocol generates the functional IPCs. Together, the encapsulation of cAD-MSC-derived IPCs and the cultivation with VSCBIC-1 enrich the maturation of generated IPCs.
Collapse
Affiliation(s)
- Quynh Dang Le
- International Program of Veterinary Science and Technology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Watchareewan Rodprasert
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Suryo Kuncorojakti
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Prasit Pavasant
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Regenerative Dentistry (CERD), Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Thanaphum Osathanon
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Chenphop Sawangmake
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.
- Department of Pharmacology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.
- Center of Excellence in Regenerative Dentistry (CERD), Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
27
|
Abstract
The ability to maintain normoglycaemia, through glucose-sensitive insulin release, is a key aspect of postnatal beta cell function. However, terminally differentiated beta cell identity does not necessarily imply functional maturity. Beta cell maturation is therefore a continuation of beta cell development, albeit a process that occurs postnatally in mammals. Although many important features have been identified in the study of beta cell maturation, as of yet no unified mechanistic model of beta cell functional maturity exists. Here, we review recent findings about the underlying mechanisms of beta cell functional maturation. These findings include systemic hormonal and nutritional triggers that operate through energy-sensing machinery shifts within beta cells, resulting in primed metabolic states that allow for appropriate glucose trafficking and, ultimately, insulin release. We also draw attention to the expansive synergistic nature of these pathways and emphasise that beta cell maturation is dependent on overlapping regulatory and metabolic networks.
Collapse
Affiliation(s)
- Tom Barsby
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Timo Otonkoski
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.
| |
Collapse
|
28
|
Abstract
The pancreatic β-cells are essential for regulating glucose homeostasis through the coordinated release of the insulin hormone. Dysfunction of the highly specialized β-cells results in diabetes mellitus, a growing global health epidemic. In this review, we describe the development and function of β-cells the emerging concept of heterogeneity within insulin-producing cells, and the potential of other cell types to assume β-cell functionality via transdifferentiation. We also discuss emerging routes to design cells with minimal β-cell properties and human stem cell differentiation efforts that carry the promise to restore normoglycemia in patients suffering from diabetes.
Collapse
Affiliation(s)
- Natanya Kerper
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA
| | - Sudipta Ashe
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
29
|
Secco B, Saitoski K, Drareni K, Soprani A, Pechberty S, Rachdi L, Venteclef N, Scharfmann R. Loss of Human Beta Cell Identity in a Reconstructed Omental Stromal Cell Environment. Cells 2022; 11:cells11060924. [PMID: 35326375 PMCID: PMC8946101 DOI: 10.3390/cells11060924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 11/30/2022] Open
Abstract
In human type 2 diabetes, adipose tissue plays an important role in disturbing glucose homeostasis by secreting factors that affect the function of cells and tissues throughout the body, including insulin-producing pancreatic beta cells. We aimed here at studying the paracrine effect of stromal cells isolated from subcutaneous and omental adipose tissue on human beta cells. We developed an in vitro model wherein the functional human beta cell line EndoC-βH1 was treated with conditioned media from human adipose tissues. By using RNA-sequencing and western blotting, we determined that a conditioned medium derived from omental stromal cells stimulates several pathways, such as STAT, SMAD and RELA, in EndoC-βH1 cells. We also observed that upon treatment, the expression of beta cell markers decreased while dedifferentiation markers increased. Loss-of-function experiments that efficiently blocked specific signaling pathways did not reverse dedifferentiation, suggesting the implication of more than one pathway in this regulatory process. Taken together, we demonstrate that soluble factors derived from stromal cells isolated from human omental adipose tissue signal human beta cells and modulate their identity.
Collapse
Affiliation(s)
- Blandine Secco
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
| | - Kevin Saitoski
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
| | - Karima Drareni
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Université de Paris, 75006 Paris, France; (K.D.); (A.S.); (N.V.)
| | - Antoine Soprani
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Université de Paris, 75006 Paris, France; (K.D.); (A.S.); (N.V.)
- Clinique Geoffroy Saint-Hilaire, Ramsey General de Santé, 75005 Paris, France
| | - Severine Pechberty
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
| | - Latif Rachdi
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
| | - Nicolas Venteclef
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Université de Paris, 75006 Paris, France; (K.D.); (A.S.); (N.V.)
| | - Raphaël Scharfmann
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
- Correspondence: ; Tel.: +(33)-1-76-53-55-68
| |
Collapse
|
30
|
Simon T, Riemer P, Jarosch A, Detjen K, Di Domenico A, Bormann F, Menne A, Khouja S, Monjé N, Childs LH, Lenze D, Leser U, Rossner F, Morkel M, Blüthgen N, Pavel M, Horst D, Capper D, Marinoni I, Perren A, Mamlouk S, Sers C. DNA methylation reveals distinct cells of origin for pancreatic neuroendocrine carcinomas and pancreatic neuroendocrine tumors. Genome Med 2022; 14:24. [PMID: 35227293 PMCID: PMC8886788 DOI: 10.1186/s13073-022-01018-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 01/28/2022] [Indexed: 02/07/2023] Open
Abstract
Background Pancreatic neuroendocrine neoplasms (PanNENs) fall into two subclasses: the well-differentiated, low- to high-grade pancreatic neuroendocrine tumors (PanNETs), and the poorly-differentiated, high-grade pancreatic neuroendocrine carcinomas (PanNECs). While recent studies suggest an endocrine descent of PanNETs, the origin of PanNECs remains unknown. Methods We performed DNA methylation analysis for 57 PanNEN samples and found that distinct methylation profiles separated PanNENs into two major groups, clearly distinguishing high-grade PanNECs from other PanNETs including high-grade NETG3. DNA alterations and immunohistochemistry of cell-type markers PDX1, ARX, and SOX9 were utilized to further characterize PanNECs and their cell of origin in the pancreas. Results Phylo-epigenetic and cell-type signature features derived from alpha, beta, acinar, and ductal adult cells suggest an exocrine cell of origin for PanNECs, thus separating them in cell lineage from other PanNENs of endocrine origin. Conclusions Our study provides a robust and clinically applicable method to clearly distinguish PanNECs from G3 PanNETs, improving patient stratification. Supplementary Information The online version contains supplementary material available at 10.1186/s13073-022-01018-w.
Collapse
Affiliation(s)
- Tincy Simon
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Pamela Riemer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Armin Jarosch
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Katharina Detjen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hepatology and Gastroenterology, Berlin, Germany
| | | | | | - Andrea Menne
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Slim Khouja
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Nanna Monjé
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Liam H Childs
- Humboldt-Universität zu Berlin, Knowledge Management in Bioinformatics, Berlin, Germany
| | - Dido Lenze
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Ulf Leser
- Humboldt-Universität zu Berlin, Knowledge Management in Bioinformatics, Berlin, Germany
| | - Florian Rossner
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Markus Morkel
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Nils Blüthgen
- Integrative Research Institute (IRI) Life Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marianne Pavel
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hepatology and Gastroenterology, Berlin, Germany
| | - David Horst
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - David Capper
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neuropathology, Berlin, Germany.,German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ilaria Marinoni
- Institute of Pathology, University of Bern, Murtenstrasse 31, 3008, Bern, Switzerland
| | - Aurel Perren
- Institute of Pathology, University of Bern, Murtenstrasse 31, 3008, Bern, Switzerland
| | - Soulafa Mamlouk
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany. .,German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Christine Sers
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany. .,German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
31
|
Mechanisms Underlying the Expansion and Functional Maturation of β-Cells in Newborns: Impact of the Nutritional Environment. Int J Mol Sci 2022; 23:ijms23042096. [PMID: 35216239 PMCID: PMC8877060 DOI: 10.3390/ijms23042096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/04/2022] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
The functional maturation of insulin-secreting β-cells is initiated before birth and is completed in early postnatal life. This process has a critical impact on the acquisition of an adequate functional β-cell mass and on the capacity to meet and adapt to insulin needs later in life. Many cellular pathways playing a role in postnatal β-cell development have already been identified. However, single-cell transcriptomic and proteomic analyses continue to reveal new players contributing to the acquisition of β-cell identity. In this review, we provide an updated picture of the mechanisms governing postnatal β-cell mass expansion and the transition of insulin-secreting cells from an immature to a mature state. We then highlight the contribution of the environment to β-cell maturation and discuss the adverse impact of an in utero and neonatal environment characterized by calorie and fat overload or by protein deficiency and undernutrition. Inappropriate nutrition early in life constitutes a risk factor for developing diabetes in adulthood and can affect the β-cells of the offspring over two generations. A better understanding of these events occurring in the neonatal period will help developing better strategies to produce functional β-cells and to design novel therapeutic approaches for the prevention and treatment of diabetes.
Collapse
|
32
|
Navik U, Rawat K, Tikoo K. L-Methionine prevents β-cell damage by modulating the expression of Arx, MafA and regulation of FOXO1 in type 1 diabetic rats. Acta Histochem 2022; 124:151820. [PMID: 34871948 DOI: 10.1016/j.acthis.2021.151820] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/31/2022]
Abstract
L-Methionine (L-Met) is an essential sulphur-containing amino acid having a vital role in various key cellular processes. Here we investigated the effect of L-Met on streptozotocin-induced β-cell damage model of diabetes mellitus in Sprague Dawley rats. At the end of study biochemical parameters, immunoblotting, qRT-PCR and ChIP-qPCR are performed. L-Met was administered orally (250 and 500 mg/kg/day) to diabetic animals for 8 weeks improved plasma glucose and insulin levels. Pancreas immunohistochemistry showed significant increase in insulin expression, decrease in glucagon and Bax expression. Interestingly, L-Met inhibited the expression of Arx; upregulated MafA and FOXO1 which play a critical role in the maintenance of β-cell identity. Our data also showed a decrease in H3K27me3 and an increase in H3K4me3 ("bivalent domain" alteration) in diabetic rats and these recovered by L-Met. Furthermore, the chromatin-immunoprecipitation assay showed a decreased enrichment of H3K27me3 on the promoter of the FOXO1 gene in diabetic rats and L-Met prevents this decrease. Our results showed the first evidence of the involvement of H3K27me3 in regulating the expression of the FOXO1 gene and the prevention of β-cell injury by L-Met treatment. In conclusion, we report the involvement of L-Met in the modulation of α-cell identity marker (Arx), β-cell identity marker (MafA) and regulation of FOXO1 by histone methylation marks for the first time. We are of the opinion that this employed as a novel therapeutic approach for mitigating diabetes-induced β-cell death.
Collapse
Affiliation(s)
- Umashanker Navik
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar (Mohali), Punjab 160062, India; Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda 151401, India.
| | - Kajal Rawat
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar (Mohali), Punjab 160062, India
| | - Kulbhushan Tikoo
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar (Mohali), Punjab 160062, India.
| |
Collapse
|
33
|
Dudek KD, Osipovich AB, Cartailler JP, Gu G, Magnuson MA. Insm1, Neurod1, and Pax6 promote murine pancreatic endocrine cell development through overlapping yet distinct RNA transcription and splicing programs. G3-GENES GENOMES GENETICS 2021; 11:6358139. [PMID: 34534285 PMCID: PMC8527475 DOI: 10.1093/g3journal/jkab303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/19/2021] [Indexed: 11/13/2022]
Abstract
Insm1, Neurod1, and Pax6 are essential for the formation and function of pancreatic endocrine cells. Here, we report comparative immunohistochemical, transcriptomic, functional enrichment, and RNA splicing analyses of these genes using gene knock-out mice. Quantitative immunohistochemical analysis confirmed that elimination of each of these three factors variably impairs the proliferation, survival, and differentiation of endocrine cells. Transcriptomic analysis revealed that each factor contributes uniquely to the transcriptome although their effects were overlapping. Functional enrichment analysis revealed that genes downregulated by the elimination of Insm1, Neurod1, and Pax6 are commonly involved in mRNA metabolism, chromatin organization, secretion, and cell cycle regulation, and upregulated genes are associated with protein degradation, autophagy, and apoptotic process. Elimination of Insm1, Neurod1, and Pax6 impaired expression of many RNA-binding proteins thereby altering RNA splicing events, including for Syt14 and Snap25, two genes required for insulin secretion. All three factors are necessary for normal splicing of Syt14, and both Insm1 and Pax6 are necessary for the processing of Snap25. Collectively, these data provide new insights into how Insm1, Neurod1, and Pax6 contribute to the formation of functional pancreatic endocrine cells.
Collapse
Affiliation(s)
- Karrie D Dudek
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA.,Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Anna B Osipovich
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | | | - Guoquing Gu
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA.,Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Mark A Magnuson
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA.,Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
34
|
Gopalan V, Singh A, Rashidi Mehrabadi F, Wang L, Ruppin E, Arda HE, Hannenhalli S. A Transcriptionally Distinct Subpopulation of Healthy Acinar Cells Exhibit Features of Pancreatic Progenitors and PDAC. Cancer Res 2021; 81:3958-3970. [PMID: 34049974 PMCID: PMC8338776 DOI: 10.1158/0008-5472.can-21-0427] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/19/2021] [Accepted: 05/26/2021] [Indexed: 12/30/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) tumors can originate either from acinar or ductal cells in the adult pancreas. We re-analyze multiple pancreas and PDAC single-cell RNA-seq datasets and find a subset of nonmalignant acinar cells, which we refer to as acinar edge (AE) cells, whose transcriptomes highly diverge from a typical acinar cell in each dataset. Genes upregulated among AE cells are enriched for transcriptomic signatures of pancreatic progenitors, acinar dedifferentiation, and several oncogenic programs. AE-upregulated genes are upregulated in human PDAC tumors, and consistently, their promoters are hypomethylated. High expression of these genes is associated with poor patient survival. The fraction of AE-like cells increases with age in healthy pancreatic tissue, which is not explained by clonal mutations, thus pointing to a nongenetic source of variation. The fraction of AE-like cells is also significantly higher in human pancreatitis samples. Finally, we find edge-like states in lung, liver, prostate, and colon tissues, suggesting that subpopulations of healthy cells across tissues can exist in pre-neoplastic states. SIGNIFICANCE: These findings show "edge" epithelial cell states with oncogenic transcriptional activity in human organs without oncogenic mutations. In the pancreas, the fraction of acinar cells increases with age.
Collapse
Affiliation(s)
- Vishaka Gopalan
- Cancer Data Science Laboratory, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland.
| | - Arashdeep Singh
- Cancer Data Science Laboratory, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Farid Rashidi Mehrabadi
- Cancer Data Science Laboratory, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
- Department of Computer Science, Indiana University, Bloomington, Indiana
| | - Li Wang
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Eytan Ruppin
- Cancer Data Science Laboratory, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - H Efsun Arda
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
35
|
Kuriyama K, Kodama Y, Shiokawa M, Nishikawa Y, Marui S, Kuwada T, Sogabe Y, Kakiuchi N, Tomono T, Matsumori T, Mima A, Morita T, Ueda T, Tsuda M, Yamauchi Y, Sakuma Y, Ota Y, Maruno T, Uza N, Kageyama R, Chiba T, Seno H. Essential role of Notch/Hes1 signaling in postnatal pancreatic exocrine development. J Gastroenterol 2021; 56:673-687. [PMID: 34128109 DOI: 10.1007/s00535-021-01779-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 03/16/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Notch/Hes1 signaling has been shown to play a role in determining the fate of pancreatic progenitor cells. However, its function in postnatal pancreatic maturation is not fully elucidated. METHODS We generated conditional Hes1 knockout and/or Notch intracellular domain (NICD) overexpression mice in Ptf1a- or Pdx1-positive pancreatic progenitor cells and analyzed pancreatic tissues. RESULTS Both Ptf1acre/+; Hes1f/f and Ptf1acre/+; Rosa26NICD mice showed normal pancreatic development at P0. However, exocrine tissue of the pancreatic tail in Ptf1acre/+; Hes1f/f mice atrophied and was replaced by fat tissue by 4 weeks of age, with increased apoptotic cells and fewer centroacinar cells. This impaired exocrine development was completely rescued by NICD overexpression in Ptf1acre/+; Hes1f/f; Rosa26NICD mice, suggesting compensation by a Notch signaling pathway other than Hes1. Conversely, Pdx1-Cre; Hes1f/f mice showed impaired postnatal exocrine development in both the pancreatic head and tail, revealing that the timing and distribution of embryonic Hes1 expression affects postnatal exocrine tissue development. CONCLUSIONS Notch signaling has an essential role in pancreatic progenitor cells for the postnatal maturation of exocrine tissue, partly through the formation of centroacinar cells.
Collapse
Affiliation(s)
- Katsutoshi Kuriyama
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yuzo Kodama
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan. .,Department of Gastroenterology, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Masahiro Shiokawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoshihiro Nishikawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Saiko Marui
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takeshi Kuwada
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yuko Sogabe
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Nobuyuki Kakiuchi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Teruko Tomono
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tomoaki Matsumori
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Atsushi Mima
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Toshihiro Morita
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tatsuki Ueda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Motoyuki Tsuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yuki Yamauchi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yojiro Sakuma
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yuji Ota
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takahisa Maruno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ryoichiro Kageyama
- Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tsutomu Chiba
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.,Kansai Electric Power Hospital, 2-1-7 Fukushima, Fukushima-ku, Osaka, 553-0003, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
36
|
Wortham M, Sander M. Transcriptional mechanisms of pancreatic β-cell maturation and functional adaptation. Trends Endocrinol Metab 2021; 32:474-487. [PMID: 34030925 PMCID: PMC8259463 DOI: 10.1016/j.tem.2021.04.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 12/31/2022]
Abstract
Pancreatic β-cells secrete insulin commensurate to circulating nutrient levels to maintain normoglycemia. The ability of β-cells to couple insulin secretion to nutrient stimuli is acquired during a postnatal maturation process. In mature β-cells the insulin secretory response adapts to changes in nutrient state. Both β-cell maturation and functional adaptation rely on the interplay between extracellular cues and cell type-specific transcriptional programs. Here we review emerging evidence that developmental and homeostatic regulation of β-cell function involves collaboration between lineage-determining and signal-dependent transcription factors (LDTFs and SDTFs, respectively). A deeper understanding of β-cell SDTFs and their cognate signals would delineate mechanisms of β-cell maturation and functional adaptation, which has direct implications for diabetes therapies and for generating mature β-cells from stem cells.
Collapse
Affiliation(s)
- Matthew Wortham
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Maike Sander
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
37
|
Milan M, Diaferia GR, Natoli G. Tumor cell heterogeneity and its transcriptional bases in pancreatic cancer: a tale of two cell types and their many variants. EMBO J 2021; 40:e107206. [PMID: 33844319 PMCID: PMC8246061 DOI: 10.15252/embj.2020107206] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), one of the most highly lethal tumors, is characterized by complex histology, with a massive fibrotic stroma in which both pseudo-glandular structures and compact nests of abnormally differentiated tumor cells are embedded, in different proportions and with different mutual relationships in space. This complexity and the heterogeneity of the tumor component have hindered the development of a broadly accepted, clinically actionable classification of PDACs, either on a morphological or a molecular basis. Here, we discuss evidence suggesting that such heterogeneity can to a large extent, albeit not exclusively, be traced back to two main classes of PDAC cells that commonly coexist in the same tumor: cells that maintained their ability to differentiate toward endodermal, mucin-producing epithelia and epithelial cells unable to form glandular structures and instead characterized by various levels of squamous differentiation and the expression of mesenchymal lineage genes. The underlying gene regulatory networks and how they are controlled by distinct transcription factors, as well as the practical implications of these two different populations of tumor cells, are discussed.
Collapse
Affiliation(s)
- Marta Milan
- Department of Experimental OncologyEuropean Institute of Oncology (IEO) IRCCSMilanItaly
- Present address:
The Francis Crick InstituteLondonUK
| | - Giuseppe R Diaferia
- Department of Experimental OncologyEuropean Institute of Oncology (IEO) IRCCSMilanItaly
| | - Gioacchino Natoli
- Department of Experimental OncologyEuropean Institute of Oncology (IEO) IRCCSMilanItaly
- Humanitas UniversityMilanItaly
| |
Collapse
|
38
|
Wu Y, Aegerter P, Nipper M, Ramjit L, Liu J, Wang P. Hippo Signaling Pathway in Pancreas Development. Front Cell Dev Biol 2021; 9:663906. [PMID: 34079799 PMCID: PMC8165189 DOI: 10.3389/fcell.2021.663906] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
The Hippo signaling pathway is a vital regulator of pancreatic development and homeostasis, directing cell fate decisions, morphogenesis, and adult pancreatic cellular plasticity. Through loss-of-function research, Hippo signaling has been found to play key roles in maintaining the proper balance between progenitor cell renewal, proliferation, and differentiation in pancreatic organogenesis. Other studies suggest that overactivation of YAP, a downstream effector of the pathway, promotes ductal cell development and suppresses endocrine cell fate specification via repression of Ngn3. After birth, disruptions in Hippo signaling have been found to lead to de-differentiation of acinar cells and pancreatitis-like phenotype. Further, Hippo signaling directs pancreatic morphogenesis by ensuring proper cell polarization and branching. Despite these findings, the mechanisms through which Hippo governs cell differentiation and pancreatic architecture are yet to be fully understood. Here, we review recent studies of Hippo functions in pancreatic development, including its crosstalk with NOTCH, WNT/β-catenin, and PI3K/Akt/mTOR signaling pathways.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States.,Department of Obstetrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Pauline Aegerter
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Michael Nipper
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Logan Ramjit
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Jun Liu
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Pei Wang
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
39
|
Almeida N, Chung MWH, Drudi EM, Engquist EN, Hamrud E, Isaacson A, Tsang VSK, Watt FM, Spagnoli FM. Employing core regulatory circuits to define cell identity. EMBO J 2021; 40:e106785. [PMID: 33934382 PMCID: PMC8126924 DOI: 10.15252/embj.2020106785] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
The interplay between extrinsic signaling and downstream gene networks controls the establishment of cell identity during development and its maintenance in adult life. Advances in next-generation sequencing and single-cell technologies have revealed additional layers of complexity in cell identity. Here, we review our current understanding of transcription factor (TF) networks as key determinants of cell identity. We discuss the concept of the core regulatory circuit as a set of TFs and interacting factors that together define the gene expression profile of the cell. We propose the core regulatory circuit as a comprehensive conceptual framework for defining cellular identity and discuss its connections to cell function in different contexts.
Collapse
Affiliation(s)
- Nathalia Almeida
- Centre for Stem Cells and Regenerative MedicineGuy’s HospitalKing’s College LondonLondonUK
| | - Matthew W H Chung
- Centre for Stem Cells and Regenerative MedicineGuy’s HospitalKing’s College LondonLondonUK
| | - Elena M Drudi
- Centre for Stem Cells and Regenerative MedicineGuy’s HospitalKing’s College LondonLondonUK
| | - Elise N Engquist
- Centre for Stem Cells and Regenerative MedicineGuy’s HospitalKing’s College LondonLondonUK
| | - Eva Hamrud
- Centre for Stem Cells and Regenerative MedicineGuy’s HospitalKing’s College LondonLondonUK
| | - Abigail Isaacson
- Centre for Stem Cells and Regenerative MedicineGuy’s HospitalKing’s College LondonLondonUK
| | - Victoria S K Tsang
- Centre for Stem Cells and Regenerative MedicineGuy’s HospitalKing’s College LondonLondonUK
| | - Fiona M Watt
- Centre for Stem Cells and Regenerative MedicineGuy’s HospitalKing’s College LondonLondonUK
| | - Francesca M Spagnoli
- Centre for Stem Cells and Regenerative MedicineGuy’s HospitalKing’s College LondonLondonUK
| |
Collapse
|
40
|
Abstract
The evolutionary origin of the nervous system has been a matter of long-standing debate. This is due to the different perspectives taken. Earlier studies addressed nervous system origins at the cellular level. They focused on the selective advantage of the first neuron in its local context, and considered vertical sensory-motor reflex arcs the first nervous system. Later studies emphasized the value of the nervous system at the tissue level. Rather than acting locally, early neurons were seen as part of an elementary nerve net that enabled the horizontal coordination of tissue movements. Opinions have also differed on the nature of effector cells. While most authors have favoured contractile systems, others see the key output of the incipient nervous system in the coordination of motile cilia, or the secretion of antimicrobial peptides. I will discuss these divergent views and explore how they can be validated by molecular and single-cell data. From this survey, possible consensus emerges: (i) the first manifestation of the nervous system likely was a nerve net, whereas specialized local circuits evolved later; (ii) different nerve nets may have evolved for the coordination of contractile or cilia-driven movements; (iii) all evolving nerve nets facilitated new forms of animal behaviour with increasing body size. This article is part of the theme issue 'Basal cognition: multicellularity, neurons and the cognitive lens'.
Collapse
Affiliation(s)
- Detlev Arendt
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Centre for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
41
|
Osipovich AB, Dudek KD, Greenfest-Allen E, Cartailler JP, Manduchi E, Potter Case L, Choi E, Chapman AG, Clayton HW, Gu G, Stoeckert CJ, Magnuson MA. A developmental lineage-based gene co-expression network for mouse pancreatic β-cells reveals a role for Zfp800 in pancreas development. Development 2021; 148:dev196964. [PMID: 33653874 PMCID: PMC8015253 DOI: 10.1242/dev.196964] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 02/17/2021] [Indexed: 12/15/2022]
Abstract
To gain a deeper understanding of pancreatic β-cell development, we used iterative weighted gene correlation network analysis to calculate a gene co-expression network (GCN) from 11 temporally and genetically defined murine cell populations. The GCN, which contained 91 distinct modules, was then used to gain three new biological insights. First, we found that the clustered protocadherin genes are differentially expressed during pancreas development. Pcdhγ genes are preferentially expressed in pancreatic endoderm, Pcdhβ genes in nascent islets, and Pcdhα genes in mature β-cells. Second, after extracting sub-networks of transcriptional regulators for each developmental stage, we identified 81 zinc finger protein (ZFP) genes that are preferentially expressed during endocrine specification and β-cell maturation. Third, we used the GCN to select three ZFPs for further analysis by CRISPR mutagenesis of mice. Zfp800 null mice exhibited early postnatal lethality, and at E18.5 their pancreata exhibited a reduced number of pancreatic endocrine cells, alterations in exocrine cell morphology, and marked changes in expression of genes involved in protein translation, hormone secretion and developmental pathways in the pancreas. Together, our results suggest that developmentally oriented GCNs have utility for gaining new insights into gene regulation during organogenesis.
Collapse
Affiliation(s)
- Anna B Osipovich
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Karrie D Dudek
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Emily Greenfest-Allen
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | - Elisabetta Manduchi
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Leah Potter Case
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Eunyoung Choi
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Austin G Chapman
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Hannah W Clayton
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Guoqiang Gu
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Christian J Stoeckert
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Mark A Magnuson
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| |
Collapse
|
42
|
Huang D, Wang R. Exploring the mechanism of pancreatic cell fate decisions via cell-cell communication. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:2401-2424. [PMID: 33892552 DOI: 10.3934/mbe.2021122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The endocrine and exocrine cells in pancreas originate initially from a group of apparently identical endoderm cells in the early gut. The endocrine and exocrine tissues are composed of islet/acinar and duct cells respectively. To explore the mechanism of pancreas cell fate decisions, we first construct a minimal mathematical model related to pancreatic regulations. The regulatory mechanism of acinar-to-islet cell conversion is revealed by bifurcation analysis of the model. In addition, Notch signaling is critical in determining the fate of endocrine and exocrine in the developing pancreas and it is a typical mediator of lateral inhibition which instructs adjacent cells to make different fate decisions. Next, we construct a multicellular model of cell-cell communication mediated by Notch signaling with trans-activation and cis-inhibition. The roles of Notch signaling in regulating fate decisions of endocrine and exocrine cells during the differentiation of pancreatic cells are explored. The results indicate that high (or low) level of Notch signaling drive cells to select the fate of exocrine (or endocrine) progenitor cells. The networks and the models presented here might be good candidates for providing qualitative mechanisms of pancreatic cell fate decisions. These results can also provide some insight on choosing perturbation strategies for further experimental analysis.
Collapse
Affiliation(s)
- Dasong Huang
- Department of Mathematics, Shanghai University, Shanghai 200444, China
| | - Ruiqi Wang
- Department of Mathematics, Shanghai University, Shanghai 200444, China
| |
Collapse
|
43
|
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
|
44
|
Balboa D, Iworima DG, Kieffer TJ. Human Pluripotent Stem Cells to Model Islet Defects in Diabetes. Front Endocrinol (Lausanne) 2021; 12:642152. [PMID: 33828531 PMCID: PMC8020750 DOI: 10.3389/fendo.2021.642152] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/03/2021] [Indexed: 12/17/2022] Open
Abstract
Diabetes mellitus is characterized by elevated levels of blood glucose and is ultimately caused by insufficient insulin production from pancreatic beta cells. Different research models have been utilized to unravel the molecular mechanisms leading to the onset of diabetes. The generation of pancreatic endocrine cells from human pluripotent stem cells constitutes an approach to study genetic defects leading to impaired beta cell development and function. Here, we review the recent progress in generating and characterizing functional stem cell-derived beta cells. We summarize the diabetes disease modeling possibilities that stem cells offer and the challenges that lie ahead to further improve these models.
Collapse
Affiliation(s)
- Diego Balboa
- Regulatory Genomics and Diabetes, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- *Correspondence: Diego Balboa,
| | - Diepiriye G. Iworima
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Timothy J. Kieffer
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
45
|
Yang J, Liu X, Yuan F, Liu J, Li D, Wei L, Wang X, Yuan L. X-box-binding protein 1 is required for pancreatic development in Xenopus laevis. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1215-1226. [PMID: 33098302 DOI: 10.1093/abbs/gmaa114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 06/25/2020] [Indexed: 11/14/2022] Open
Abstract
X-box-binding protein 1 (XBP1) is a protein containing the basic leucine zipper structure. It belongs to the cAMP-response element binding protein (CREB)/activating transcription factor transcription factor family. As the main transcription factor, spliced XBP1 (XBP1s) participates in many physiological and pathological processes and plays an important role in embryonic development. Previous studies showed that XBP1-knockout mice died because of pancreatic exocrine function deficiency, indicating that XBP1 plays an important role in pancreatic development. However, the exact role of XBP1 in pancreatic development remains unclear. This study aimed to investigate the role of XBP1 in the pancreatic development of Xenopus laevis embryos. Whole-mount in situ hybridization and quantitative real-time PCR results revealed that the expression levels of pancreatic progenitor marker genes pdx1, p48, ngn3, and sox9 were downregulated in XBP1s morpholino oligonucleotide (MO)-injected embryos. The expression levels of pancreatic exocrine and endocrine marker genes insulin and amylase were also downregulated. Through the overexpression of XBP1s, the phenotype and gene expressions were opposite to those in XBP1s MO-injected embryos. Luciferase and chromatin immunoprecipitation assays showed that XBP1s could bind to the XBP1-binding site in the foxa2 promoter. These results revealed that XBP1 is required in the pancreatic development of Xenopus laevis and might function by regulating foxa2.
Collapse
Affiliation(s)
- Jing Yang
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing 210029, China
| | - Xingjing Liu
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, Medical School, Southeast University, Nanjing 210009, China
| | - Fang Yuan
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing 210029, China
| | - Jia Liu
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing 210029, China
| | - Deli Li
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing 210029, China
| | - Liyuan Wei
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing 210029, China
| | - Xuejun Wang
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing 210029, China
| | - Li Yuan
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing 210029, China
| |
Collapse
|
46
|
Baumann D, Wong A, Akhaphong B, Jo S, Pritchard S, Mohan R, Chung G, Zhang Y, Alejandro EU. Role of nutrient-driven O-GlcNAc-post-translational modification in pancreatic exocrine and endocrine islet development. Development 2020; 147:dev186643. [PMID: 32165492 PMCID: PMC7174839 DOI: 10.1242/dev.186643] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/25/2020] [Indexed: 12/16/2022]
Abstract
Although the developing pancreas is exquisitely sensitive to nutrient supply in utero, it is not entirely clear how nutrient-driven post-translational modification of proteins impacts the pancreas during development. We hypothesized that the nutrient-sensing enzyme O-GlcNAc transferase (Ogt), which catalyzes an O-GlcNAc-modification onto key target proteins, integrates nutrient-signaling networks to regulate cell survival and development. In this study, we investigated the heretofore unknown role of Ogt in exocrine and endocrine islet development. By genetic manipulation in vivo and by using morphometric and molecular analyses, such as immunofluorescence imaging and single cell RNA sequencing, we show the first evidence that Ogt regulates pancreas development. Genetic deletion of Ogt in the pancreatic epithelium (OgtKOPanc) causes pancreatic hypoplasia, in part by increased apoptosis and reduced levels of of Pdx1 protein. Transcriptomic analysis of single cell and bulk RNA sequencing uncovered cell-type heterogeneity and predicted upstream regulator proteins that mediate cell survival, including Pdx1, Ptf1a and p53, which are putative Ogt targets. In conclusion, these findings underscore the requirement of O-GlcNAcylation during pancreas development and show that Ogt is essential for pancreatic progenitor survival, providing a novel mechanistic link between nutrients and pancreas development.
Collapse
Affiliation(s)
- Daniel Baumann
- Department of Integrative Biology and Physiology, University of Minnesota Medical School. Minneapolis, MN 55455, USA
| | - Alicia Wong
- Department of Integrative Biology and Physiology, University of Minnesota Medical School. Minneapolis, MN 55455, USA
| | - Brian Akhaphong
- Department of Integrative Biology and Physiology, University of Minnesota Medical School. Minneapolis, MN 55455, USA
| | - Seokwon Jo
- Department of Integrative Biology and Physiology, University of Minnesota Medical School. Minneapolis, MN 55455, USA
| | - Samantha Pritchard
- Department of Integrative Biology and Physiology, University of Minnesota Medical School. Minneapolis, MN 55455, USA
| | - Ramkumar Mohan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School. Minneapolis, MN 55455, USA
| | - Grace Chung
- Department of Integrative Biology and Physiology, University of Minnesota Medical School. Minneapolis, MN 55455, USA
| | - Ying Zhang
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Emilyn U Alejandro
- Department of Integrative Biology and Physiology, University of Minnesota Medical School. Minneapolis, MN 55455, USA
| |
Collapse
|
47
|
Niu N, Lu P, Yang Y, He R, Zhang L, Shi J, Wu J, Yang M, Zhang ZG, Wang LW, Gao WQ, Habtezion A, Xiao GG, Sun Y, Li L, Xue J. Loss of Setd2 promotes Kras-induced acinar-to-ductal metaplasia and epithelia-mesenchymal transition during pancreatic carcinogenesis. Gut 2020; 69:715-726. [PMID: 31300513 DOI: 10.1136/gutjnl-2019-318362] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/18/2019] [Accepted: 06/30/2019] [Indexed: 01/04/2023]
Abstract
OBJECTIVE SETD2, the sole histone H3K36 trimethyltransferase, is frequently mutated or deleted in human cancer, including pancreatic ductal adenocarcinoma (PDAC). However, whether SETD2/H3K36me3 alteration results in PDAC remains largely unknown. DESIGN TCGA(PAAD) public database and PDAC tissue array with SETD2/H3K36me3 staining were used to investigate the clinical relevance of SETD2 in PDAC. Furthermore, to define the role of SETD2 in the carcinogenesis of PDAC, we crossed conditional Setd2 knockout mice (PdxcreSetd2flox/flox) together with KrasG12D mice. Moreover, to examine the role of SETD2 after ductal metaplasia, Crisp/cas9 was used to deplete Setd2 in PDAC cells. RNA-seq and H3K36me3 ChIP-seq were performed to uncover the mechanism. RESULTS SETD2 mutant/low expression was correlated with poor prognosis in patients with PDAC. Next, we found that Setd2 acted as a putative tumour suppressor in Kras-driven pancreatic carcinogenesis. Mechanistically, Setd2 loss in acinar cells facilitated Kras-induced acinar-to-ductal reprogramming, mainly through epigenetic dysregulation of Fbxw7. Moreover, Setd2 ablation in pancreatic cancer cells enhanced epithelia-mesenchymal transition (EMT) through impaired epigenetic regulation of Ctnna1. In addition, Setd2 deficiency led to sustained Akt activation via inherent extracellular matrix (ECM) production, which would favour their metastasis. CONCLUSION Together, our findings highlight the function of SETD2 during pancreatic carcinogenesis, which would advance our understanding of epigenetic dysregulation in PDAC. Moreover, it may also pave the way for development of targeted, patients-tailored therapies for PDAC patients with SETD2 deficiency.
Collapse
Affiliation(s)
- Ningning Niu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Ping Lu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Yanlin Yang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Ruizhe He
- Department of Biliary-Pancreatic Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Juanjuan Shi
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Jinghua Wu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Minwei Yang
- Department of Biliary-Pancreatic Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-Wei Wang
- Department of Oncology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China.,School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Aida Habtezion
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California, USA
| | - Gary Guishan Xiao
- School of pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China
| | - Yongwei Sun
- Department of Biliary-Pancreatic Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Li Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China.,School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Xue
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| |
Collapse
|
48
|
Kim S, Whitener RL, Peiris H, Gu X, Chang CA, Lam JY, Camunas-Soler J, Park I, Bevacqua RJ, Tellez K, Quake SR, Lakey JRT, Bottino R, Ross PJ, Kim SK. Molecular and genetic regulation of pig pancreatic islet cell development. Development 2020; 147:dev186213. [PMID: 32108026 PMCID: PMC7132804 DOI: 10.1242/dev.186213] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 02/20/2020] [Indexed: 12/12/2022]
Abstract
Reliance on rodents for understanding pancreatic genetics, development and islet function could limit progress in developing interventions for human diseases such as diabetes mellitus. Similarities of pancreas morphology and function suggest that porcine and human pancreas developmental biology may have useful homologies. However, little is known about pig pancreas development. To fill this knowledge gap, we investigated fetal and neonatal pig pancreas at multiple, crucial developmental stages using modern experimental approaches. Purification of islet β-, α- and δ-cells followed by transcriptome analysis (RNA-seq) and immunohistology identified cell- and stage-specific regulation, and revealed that pig and human islet cells share characteristic features that are not observed in mice. Morphometric analysis also revealed endocrine cell allocation and architectural similarities between pig and human islets. Our analysis unveiled scores of signaling pathways linked to native islet β-cell functional maturation, including evidence of fetal α-cell GLP-1 production and signaling to β-cells. Thus, the findings and resources detailed here show how pig pancreatic islet studies complement other systems for understanding the developmental programs that generate functional islet cells, and that are relevant to human pancreatic diseases.
Collapse
Affiliation(s)
- Seokho Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Robert L Whitener
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Heshan Peiris
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xueying Gu
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Charles A Chang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jonathan Y Lam
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joan Camunas-Soler
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Insung Park
- Department of Animal Science, University of California Davis, Davis, CA 95616, USA
| | - Romina J Bevacqua
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Krissie Tellez
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94518, USA
| | - Jonathan R T Lakey
- Department of Surgery, University of California at Irvine, Irvine, CA 92868, USA
| | - Rita Bottino
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA 15212, USA
| | - Pablo J Ross
- Department of Animal Science, University of California Davis, Davis, CA 95616, USA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
49
|
Wang J, Yuan R, Zhu X, Ao P. Adaptive Landscape Shaped by Core Endogenous Network Coordinates Complex Early Progenitor Fate Commitments in Embryonic Pancreas. Sci Rep 2020; 10:1112. [PMID: 31980678 PMCID: PMC6981170 DOI: 10.1038/s41598-020-57903-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 12/07/2019] [Indexed: 02/06/2023] Open
Abstract
The classical development hierarchy of pancreatic cell fate commitments describes that multipotent progenitors (MPs) first bifurcate into tip cells and trunk cells, and then these cells give rise to acinar cells and endocrine/ductal cells separately. However, lineage tracings reveal that pancreatic progenitors are highly heterogeneous in tip and trunk domains in embryonic pancreas. The progenitor fate commitments from multipotency to unipotency during early pancreas development is insufficiently characterized. In pursuing a mechanistic understanding of the complexity in progenitor fate commitments, we construct a core endogenous network for pancreatic lineage decisions based on genetic regulations and quantified its intrinsic dynamic properties using dynamic modeling. The dynamics reveal a developmental landscape with high complexity that has not been clarified. Not only well-characterized pancreatic cells are reproduced, but also previously unrecognized progenitors-tip progenitor (TiP), trunk progenitor (TrP), later endocrine progenitor (LEP), and acinar progenitors (AciP/AciP2) are predicted. Further analyses show that TrP and LEP mediate endocrine lineage maturation, while TiP, AciP, AciP2 and TrP mediate acinar and ductal lineage maturation. The predicted cell fate commitments are validated by analyzing single-cell RNA sequencing (scRNA-seq) data. Significantly, this is the first time that a redefined hierarchy with detailed early pancreatic progenitor fate commitment is obtained.
Collapse
Affiliation(s)
- Junqiang Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruoshi Yuan
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaomei Zhu
- Shanghai Center for Quantitative Life Sciences and Physics Department, Shanghai University, Shanghai, China
| | - Ping Ao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Center for Quantitative Life Sciences and Physics Department, Shanghai University, Shanghai, China.
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
- State Key Laboratory for Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
50
|
Salinno C, Cota P, Bastidas-Ponce A, Tarquis-Medina M, Lickert H, Bakhti M. β-Cell Maturation and Identity in Health and Disease. Int J Mol Sci 2019; 20:E5417. [PMID: 31671683 PMCID: PMC6861993 DOI: 10.3390/ijms20215417] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022] Open
Abstract
The exponential increase of patients with diabetes mellitus urges for novel therapeutic strategies to reduce the socioeconomic burden of this disease. The loss or dysfunction of insulin-producing β-cells, in patients with type 1 and type 2 diabetes respectively, put these cells at the center of the disease initiation and progression. Therefore, major efforts have been taken to restore the β-cell mass by cell-replacement or regeneration approaches. Implementing novel therapies requires deciphering the developmental mechanisms that generate β-cells and determine the acquisition of their physiological phenotype. In this review, we summarize the current understanding of the mechanisms that coordinate the postnatal maturation of β-cells and define their functional identity. Furthermore, we discuss different routes by which β-cells lose their features and functionality in type 1 and 2 diabetic conditions. We then focus on potential mechanisms to restore the functionality of those β-cell populations that have lost their functional phenotype. Finally, we discuss the recent progress and remaining challenges facing the generation of functional mature β-cells from stem cells for cell-replacement therapy for diabetes treatment.
Collapse
Affiliation(s)
- Ciro Salinno
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.
- School of Medicine, Technical University of Munich, 81675Munich, Germany.
| | - Perla Cota
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.
- School of Medicine, Technical University of Munich, 81675Munich, Germany.
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.
- School of Medicine, Technical University of Munich, 81675Munich, Germany.
| | - Marta Tarquis-Medina
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.
- School of Medicine, Technical University of Munich, 81675Munich, Germany.
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.
- School of Medicine, Technical University of Munich, 81675Munich, Germany.
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.
| |
Collapse
|