1
|
Karakose E, Wang X, Wang P, Carcamo S, Demircioglu D, Lambertini L, Wood O, Kang R, Lu G, Scott DK, Garcia-Ocaña A, Argmann C, Sebra RP, Hasson D, Stewart AF. Cycling alpha cells in regenerative drug-treated human pancreatic islets may serve as key beta cell progenitors. Cell Rep Med 2024; 5:101832. [PMID: 39626675 PMCID: PMC11722108 DOI: 10.1016/j.xcrm.2024.101832] [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: 11/24/2023] [Revised: 07/30/2024] [Accepted: 10/30/2024] [Indexed: 12/20/2024]
Abstract
Diabetes results from an inadequate number of insulin-producing human beta cells. There is currently no clinically available effective means to restore beta cell mass in millions of people with diabetes. Although the DYRK1A inhibitors, either alone or in combination with GLP-1 receptor agonists (GLP-1) or transforming growth factor β (TGF-β) superfamily inhibitors (LY), induce beta cell replication and increase beta cell mass, the precise mechanisms of action remain elusive. Here we perform single-cell RNA sequencing on human pancreatic islets treated with a DYRK1A inhibitor, either alone or with GLP-1 or LY. We identify cycling alpha cells as the most responsive cells to DYRK1A inhibition. Lineage trajectory analyses suggest that cycling alpha cells may serve as precursor cells that transdifferentiate into beta cells. Collectively, in addition to enhancing expression of beta cell phenotypic genes in beta cells, our findings suggest that regenerative drugs may be targeting cycling alpha cells in human islets.
Collapse
Affiliation(s)
- Esra Karakose
- Diabetes, Obesity, Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Xuedi Wang
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) Core, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Peng Wang
- Diabetes, Obesity, Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Saul Carcamo
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) Core, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Deniz Demircioglu
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) Core, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Luca Lambertini
- Diabetes, Obesity, Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Olivia Wood
- Diabetes, Obesity, Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Randy Kang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Geming Lu
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Donald K Scott
- Diabetes, Obesity, Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adolfo Garcia-Ocaña
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carmen Argmann
- Department of Genetics and Genomic Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert P Sebra
- Department of Genetics and Genomic Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dan Hasson
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) Core, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrew F Stewart
- Diabetes, Obesity, Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
2
|
Chen Y, Yin X, Xu R, Ruze R, Song J, Yin C, Hu C, Wang C, Xu Q, Zhao Y. Cancer-Associated Endocrine Cells Participate in Pancreatic Carcinogenesis. Gastroenterology 2024; 167:1167-1182.e23. [PMID: 39048054 DOI: 10.1053/j.gastro.2024.07.016] [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: 12/12/2023] [Revised: 06/20/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND & AIMS The pancreas is composed of endocrine and exocrine parts, and its interlacing structure indicates potential interaction between endocrine and exocrine cells. Although the tumor microenvironment of pancreatic ductal adenocarcinoma (PDAC) has been well characterized, the role of pancreatic endocrine cells during carcinogenesis is relatively understudied. METHODS The changes of endocrine cells in PDAC by single-cell transcriptome sequencing, spatial transcriptome sequencing, and multiplex immunohistochemistry were depicted. After that, the interaction between pancreatic carcinogenesis and endocrine changes was explored in orthotopic transplantation mice, KrasLSL-G12DPdx1-Cre mice, and KrasLSL-G12Dp53LoxPPdx1-CreER mice. Finally, we proved the mechanism of the interaction between endocrine and exocrine parts of the pancreas through islet isolation, co-culture in vitro and co-injection in vivo. RESULTS Pancreatic endocrine cells displayed significantly different transcriptomic characteristics and increased interaction with exocrine part in PDAC. Specifically, among all of the changes, pancreatic polypeptide-positive cells showed a sharp increment accompanied by the progression of the cancer lesion, which might be derived from the transdifferentiation of α and β cells. Interestingly, it was proved that PDAC cells were able to induce the transdifferentiation of pancreatic α cells and β cells into glucagon-pancreatic polypeptide and insulin-pancreatic polypeptide double-positive cells, which further promoted carcinogenesis and development of PDAC in a paracrine-dependent manner and formed a reciprocal interaction. CONCLUSIONS This study systematically maps the alteration of pancreatic endocrine cells in PDAC and elucidates the potential endocrine-exocrine interaction mechanisms during PDAC carcinogenesis. In addition, cancer-associated endocrine cells are defined and characterized, thereby further broadening the composition of PDAC microenvironment.
Collapse
MESH Headings
- Animals
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Mice
- Humans
- Tumor Microenvironment
- Coculture Techniques
- Single-Cell Analysis
- Cell Transdifferentiation
- Insulin-Secreting Cells/pathology
- Insulin-Secreting Cells/metabolism
- Transcriptome
- Cell Line, Tumor
- Glucagon-Secreting Cells/pathology
- Glucagon-Secreting Cells/metabolism
- Carcinogenesis/pathology
- Carcinogenesis/genetics
- Gene Expression Regulation, Neoplastic
- Cell Transformation, Neoplastic/pathology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/genetics
- Gene Expression Profiling
- Disease Models, Animal
- Mice, Transgenic
Collapse
Affiliation(s)
- Yuan Chen
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Xinpeng Yin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Rexiati Ruze
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Jianlu Song
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Chenxue Yin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Chenglin Hu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Chengcheng Wang
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China; National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, People's Republic of China; Institute of Clinical Medicine, Peking Union Medical College Hospital, Beijing, People's Republic of China.
| | - Qiang Xu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China; National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, People's Republic of China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, People's Republic of China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China; National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, People's Republic of China.
| |
Collapse
|
3
|
Chen Y, Jiang Q, Xing X, Yuan T, Li P. Clinical research progress on β-cell dysfunction in T2DM development in the Chinese population. Rev Endocr Metab Disord 2024:10.1007/s11154-024-09914-9. [PMID: 39382753 DOI: 10.1007/s11154-024-09914-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/06/2024] [Indexed: 10/10/2024]
Abstract
The prevalence of type-2 diabetes mellitus (T2DM) has increased over 10-fold in the past 40 years in China, which now has the largest T2DM population in the world. Insulin resistance and β-cell dysfunction are the typical features of T2DM. Although both factors play a role, decreased β-cell function and β-cell mass are the predominant factors for progression to T2DM. Considering the differences between Chinese T2DM patients and those of other ethnicities, it is important to characterize β-cell dysfunction in Chinese patients during T2DM progression. Herein, we reviewed the studies on the relationships between β-cell function and T2DM progression in the Chinese population and discussed the differences among individuals of varying ethnicities. Meanwhile, we summarized the risk factors and current treatments of T2DM in Chinese individuals and discussed their impacts on β-cell function with the hope of identifying a better T2DM therapy.
Collapse
Affiliation(s)
- Yibing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, 100050, China
| | - Qian Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, 100050, China
| | - Xiaowei Xing
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, 100050, China
| | - Tao Yuan
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Pingping Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, 100050, China.
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, 100050, China.
| |
Collapse
|
4
|
Cobo-Vuilleumier N, Lorenzo PI, Martin Vazquez E, López Noriega L, Nano R, Piemonti L, Martín F, Gauthier BR. Enhancing human islet xenotransplant survival and function in diabetic immunocompetent mice through LRH-1/NR5A2 pharmacological activation. Front Immunol 2024; 15:1470881. [PMID: 39399499 PMCID: PMC11466778 DOI: 10.3389/fimmu.2024.1470881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
The intricate etiology of type 1 diabetes mellitus (T1D), characterized by harmful interactions between the immune system and insulin-producing beta cells, has hindered the development of effective therapies including human islet transplantation, which requires strong immunosuppressants that impair beta cell survival and function. As such alternative immunomodulating therapies are required for successful transplantation. The discovery that pharmacological activation of the nuclear receptor LRH-1/NR5A2 can reverse hyperglycemia in mouse models of T1D by altering, and not suppressing the autoimmune attack, prompted us to investigate whether LRH-1/NR5A2 activation could improve human islet function/survival after xenotransplantation in immunocompetent mice. Human islets were transplanted under the kidney capsule of streptozotocin (STZ)-induced diabetic mice, and treatment with BL001 (LRH-1/NR5A2 agonist) or vehicle was administered one week post-transplant. Our study, encompassing 3 independent experiments with 3 different islet donors, revealed that mice treated for 8 weeks with BL001 exhibited lower blood glucose levels correlating with improved mouse survival rates as compared to vehicle-treated controls. Human C-peptide was detectable in BL001-treated mice at both 4 and 8 weeks indicating functional islet beta cells. Accordingly, in mice treated with BL001 for 8 weeks, the beta cell mass was preserved, while a significant decrease in alpha cells was observed compared to mice treated with BL001 for only 4 weeks. In contrast, vehicle-treated mice exhibited a reduction in insulin-expressing cells at 8 weeks compared to those at 4 weeks. These results suggest that BL001 significantly enhances the survival, engraftment, and functionality of human islets in a STZ-induced diabetic mouse model.
Collapse
Affiliation(s)
- N. Cobo-Vuilleumier
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
| | - P. I. Lorenzo
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
| | - E. Martin Vazquez
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
| | - L. López Noriega
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
| | - R. Nano
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - L. Piemonti
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - F. Martín
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - B. R. Gauthier
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| |
Collapse
|
5
|
Staels W, Berthault C, Bourgeois S, Laville V, Lourenço C, De Leu N, Scharfmann R. Comprehensive alpha, beta, and delta cell transcriptomics reveal an association of cellular aging with MHC class I upregulation. Mol Metab 2024; 87:101990. [PMID: 39009220 PMCID: PMC11327396 DOI: 10.1016/j.molmet.2024.101990] [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: 05/08/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024] Open
Abstract
OBJECTIVES This study aimed to evaluate the efficacy of a purification method developed for isolating alpha, beta, and delta cells from pancreatic islets of adult mice, extending its application to islets from newborn and aged mice. Furthermore, it sought to examine transcriptome dynamics in mouse pancreatic endocrine islet cells throughout postnatal development and to validate age-related alterations within these cell populations. METHODS We leveraged the high surface expression of CD71 on beta cells and CD24 on delta cells to FACS-purify alpha, beta, and delta cells from newborn (1-week-old), adult (12-week-old), and old (18-month-old) mice. Bulk RNA sequencing was conducted on these purified cell populations, and subsequent bioinformatic analyses included differential gene expression, overrepresentation, and intersection analysis. RESULTS Alpha, beta, and delta cells from newborn and aged mice were successfully FACS-purified using the same method employed for adult mice. Our analysis of the age-related transcriptional changes in alpha, beta, and delta cell populations revealed a decrease in cell cycling and an increase in neuron-like features processes during the transition from newborn to adult mice. Progressing from adult to old mice, we identified an inflammatory gene signature related to aging (inflammaging) encompassing an increase in β-2 microglobulin and major histocompatibility complex (MHC) Class I expression. CONCLUSIONS Our study demonstrates the effectiveness of our cell sorting technique in purifying endocrine subsets from mouse islets at different ages. We provide a valuable resource for better understanding endocrine pancreas aging and identified an inflammaging gene signature with increased β-2 microglobulin and MHC Class I expression as a common hallmark of old alpha, beta, and delta cells, with potential implications for immune response regulation and age-related diabetes.
Collapse
Affiliation(s)
- W Staels
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France; Genetics, Reproduction and Development (GRAD), Vrije Universiteit Brussel (VUB), Brussels, Belgium; Division of Pediatric Endocrinology, Department of Pediatrics, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium.
| | - C Berthault
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - S Bourgeois
- Genetics, Reproduction and Development (GRAD), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - V Laville
- Stem Cells and Development Unit, Institut Pasteur, Paris, France; UMR CNRS 3738, Institut Pasteur, Paris, France; Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - C Lourenço
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - N De Leu
- Genetics, Reproduction and Development (GRAD), Vrije Universiteit Brussel (VUB), Brussels, Belgium; Endocrinology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium; Endocrinology, ASZ Aalst, 9300 Aalst, Belgium
| | - R Scharfmann
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France
| |
Collapse
|
6
|
Toledo MP, Xie G, Wang YJ. Comprehensive Characterization of Islet Remodeling in Development and in Diabetes Using Mass Cytometry. Endocrinology 2024; 165:bqae094. [PMID: 39058908 DOI: 10.1210/endocr/bqae094] [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: 04/30/2024] [Revised: 07/11/2024] [Accepted: 07/25/2024] [Indexed: 07/28/2024]
Abstract
The pancreatic islet is the functional and structural unit of the pancreatic endocrine portion. Islet remodeling occurs in both normal development and pathogenesis of type 1 (T1D) and type 2 diabetes (T2D). However, accurately quantifying changes in islet cellular makeup and hormone expressions poses significant challenges due to large intra- and inter-donor heterogeneity and the limited scalability of traditional methods such as immunostaining. The cytometry by time-of-flight (CyTOF) technology enables simultaneous quantification of more than 30 protein markers at single-cell resolution in a high-throughput fashion. Moreover, with distinct DNA and viability markers, single live cells can be explicitly selected in CyTOF. Here, leveraging the CyTOF data generated by the Human Pancreas Analysis Program, we characterized more than 12 million islet cells from 71 donors. Our data revealed continued age-related changes in islet endocrine cell compositions, but the maturity of endocrine cells is reached by 3 years of age. We also observed significant changes in beta cell numbers and key protein expressions, along with a significant increase in bihormonal cells in T1D donors. In contrast, T2D donors exhibited minimal islet remodeling events. Our data shine a light on the islet dynamics during development and diabetes pathogenesis and suggest divergent pathogenesis processes of T1D and T2D. Our comprehensive approach not only elucidates islet plasticity but also establishes a foundation for integrated CyTOF analysis in islet biology and beyond.
Collapse
Affiliation(s)
- Maria Pilar Toledo
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Gengqiang Xie
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Yue J Wang
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
7
|
Ampofo E, Pack M, Wrublewsky S, Boewe AS, Spigelman AF, Koch H, MacDonald PE, Laschke MW, Montenarh M, Götz C. CK2 activity is crucial for proper glucagon expression. Diabetologia 2024; 67:1368-1385. [PMID: 38503901 PMCID: PMC11153270 DOI: 10.1007/s00125-024-06128-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/07/2024] [Indexed: 03/21/2024]
Abstract
AIMS/HYPOTHESIS Protein kinase CK2 acts as a negative regulator of insulin expression in pancreatic beta cells. This action is mainly mediated by phosphorylation of the transcription factor pancreatic and duodenal homeobox protein 1 (PDX1). In pancreatic alpha cells, PDX1 acts in a reciprocal fashion on glucagon (GCG) expression. Therefore, we hypothesised that CK2 might positively regulate GCG expression in pancreatic alpha cells. METHODS We suppressed CK2 kinase activity in αTC1 cells by two pharmacological inhibitors and by the CRISPR/Cas9 technique. Subsequently, we analysed GCG expression and secretion by real-time quantitative RT-PCR, western blot, luciferase assay, ELISA and DNA pull-down assays. We additionally studied paracrine effects on GCG secretion in pseudoislets, isolated murine islets and human islets. In vivo, we examined the effect of CK2 inhibition on blood glucose levels by systemic and alpha cell-specific CK2 inhibition. RESULTS We found that CK2 downregulation reduces GCG secretion in the murine alpha cell line αTC1 (e.g. from 1094±124 ng/l to 459±110 ng/l) by the use of the CK2-inhibitor SGC-CK2-1. This was due to a marked decrease in Gcg gene expression through alteration of the binding of paired box protein 6 (PAX6) and transcription factor MafB to the Gcg promoter. The analysis of the underlying mechanisms revealed that both transcription factors are displaced by PDX1. Ex vivo experiments in isolated murine islets and pseudoislets further demonstrated that CK2-mediated reduction in GCG secretion was only slightly affected by the higher insulin secretion after CK2 inhibition. The kidney capsule transplantation model showed the significance of CK2 for GCG expression and secretion in vivo. Finally, CK2 downregulation also reduced the GCG secretion in islets isolated from humans. CONCLUSIONS/INTERPRETATION These novel findings not only indicate an important function of protein kinase CK2 for proper GCG expression but also demonstrate that CK2 may be a promising target for the development of novel glucose-lowering drugs.
Collapse
Affiliation(s)
- Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Mandy Pack
- Medical Biochemistry and Molecular Biology, Saarland University, Homburg, Germany
| | - Selina Wrublewsky
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Anne S Boewe
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Aliya F Spigelman
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Hanna Koch
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Patrick E MacDonald
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Mathias Montenarh
- Medical Biochemistry and Molecular Biology, Saarland University, Homburg, Germany
| | - Claudia Götz
- Medical Biochemistry and Molecular Biology, Saarland University, Homburg, Germany.
| |
Collapse
|
8
|
Choi J, Cayabyab F, Perez H, Yoshihara E. Scaling Insulin-Producing Cells by Multiple Strategies. Endocrinol Metab (Seoul) 2024; 39:191-205. [PMID: 38572534 PMCID: PMC11066437 DOI: 10.3803/enm.2023.1910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/20/2024] [Accepted: 01/30/2024] [Indexed: 04/05/2024] Open
Abstract
In the quest to combat insulin-dependent diabetes mellitus (IDDM), allogenic pancreatic islet cell therapy sourced from deceased donors represents a significant therapeutic advance. However, the applicability of this approach is hampered by donor scarcity and the demand for sustained immunosuppression. Human induced pluripotent stem cells are a game-changing resource for generating synthetic functional insulin-producing β cells. In addition, novel methodologies allow the direct expansion of pancreatic progenitors and mature β cells, thereby circumventing prolonged differentiation. Nevertheless, achieving practical reproducibility and scalability presents a substantial challenge for this technology. As these innovative approaches become more prominent, it is crucial to thoroughly evaluate existing expansion techniques with an emphasis on their optimization and scalability. This manuscript delineates these cutting-edge advancements, offers a critical analysis of the prevailing strategies, and underscores pivotal challenges, including cost-efficiency and logistical issues. Our insights provide a roadmap, elucidating both the promises and the imperatives in harnessing the potential of these cellular therapies for IDDM.
Collapse
Affiliation(s)
- Jinhyuk Choi
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Fritz Cayabyab
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Harvey Perez
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Eiji Yoshihara
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
9
|
Puri S, Maachi H, Nair G, Russ HA, Chen R, Pulimeno P, Cutts Z, Ntranos V, Hebrok M. Sox9 regulates alternative splicing and pancreatic beta cell function. Nat Commun 2024; 15:588. [PMID: 38238288 PMCID: PMC10796970 DOI: 10.1038/s41467-023-44384-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
Despite significant research, mechanisms underlying the failure of islet beta cells that result in type 2 diabetes (T2D) are still under investigation. Here, we report that Sox9, a transcriptional regulator of pancreas development, also functions in mature beta cells. Our results show that Sox9-depleted rodent beta cells have defective insulin secretion, and aging animals develop glucose intolerance, mimicking the progressive degeneration observed in T2D. Using genome editing in human stem cells, we show that beta cells lacking SOX9 have stunted first-phase insulin secretion. In human and rodent cells, loss of Sox9 disrupts alternative splicing and triggers accumulation of non-functional isoforms of genes with key roles in beta cell function. Sox9 depletion reduces expression of protein-coding splice variants of the serine-rich splicing factor arginine SRSF5, a major splicing enhancer that regulates alternative splicing. Our data highlight the role of SOX9 as a regulator of alternative splicing in mature beta cell function.
Collapse
Affiliation(s)
- Sapna Puri
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA
- Minutia Inc., Oakland, CA, USA
| | - Hasna Maachi
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA
- Center for Organoid Systems, Klinikum Rechts der Isar (MRI) and Technical University Munich, 85748, Garching, Germany
- Institute for Diabetes Organoid Technology, Helmholtz Munich, Helmholtz Diabetes Center, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Munich Institute of Biomedical Engineering (MIBE), Technical University Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany
| | - Gopika Nair
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA
- Eli Lilly, Indianapolis, IN, USA
| | - Holger A Russ
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA
- Diabetes Institute, University of Florida, Gainesville, FL, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Richard Chen
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA
| | - Pamela Pulimeno
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA
| | - Zachary Cutts
- Graduate Program in Bioinformatics, University of California, San Francisco, CA, USA
| | - Vasilis Ntranos
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA, USA.
- Center for Organoid Systems, Klinikum Rechts der Isar (MRI) and Technical University Munich, 85748, Garching, Germany.
- Institute for Diabetes Organoid Technology, Helmholtz Munich, Helmholtz Diabetes Center, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
- Munich Institute of Biomedical Engineering (MIBE), Technical University Munich, Munich, Germany.
- German Center for Diabetes Research (DZD), Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany.
| |
Collapse
|
10
|
Leung SS, Lenchik N, Mathews C, Pugliese A, McCarthy DA, Le Bagge S, Ewing A, Harris M, Radford KJ, Borg DJ, Gerling I, Forbes JM. Alpha cell receptor for advanced glycation end products associate with glucagon expression in type 1 diabetes. Sci Rep 2023; 13:12948. [PMID: 37558746 PMCID: PMC10412557 DOI: 10.1038/s41598-023-39243-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: 05/08/2023] [Accepted: 07/21/2023] [Indexed: 08/11/2023] Open
Abstract
Hypoglycemia in type 1 diabetes associates with changes in the pancreatic islet α cells, where the receptor for advanced glycation end products (RAGE) is highly expressed. This study compared islet RAGE expression in donors without diabetes, those at risk of, and those with type 1 diabetes. Laser-dissected islets were subject to RNA bioinformatics and adjacent pancreatic tissue were assessed by confocal microscopy. We found that islets from type 1 diabetes donors had differential expression of the RAGE gene (AGER) and its correlated genes, based on glucagon expression. Random forest machine learning revealed that AGER was the most important predictor for islet glucagon levels. Conversely, a generalized linear model identified that glucagon expression could be predicted by expression of RAGE signaling molecules, its ligands and enzymes that create or clear RAGE ligands. Confocal imaging co-localized RAGE, its ligands and signaling molecules to the α cells. Half of the type 1 diabetes cohort comprised of adolescents and a patient with history of hypoglycemia-all showed an inverse relationship between glucagon and RAGE. These data confirm an association between glucagon and islet RAGE, its ligands and signaling pathways in type 1 diabetes, which warrants functional investigation into a role for RAGE in hypoglycemia.
Collapse
Affiliation(s)
- Sherman S Leung
- Glycation and Diabetes Complications, Mater Research Institute, Translational Research Institute (TRI), The University of Queensland (MRI-UQ), 37 Kent Street, Woolloongabba, Brisbane, QLD, 4102, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- School of Medicine and Dentistry, Griffith University, Brisbane, Australia
- Wesley Research Institute, The Wesley Hospital, Brisbane, Australia
| | - Nataliya Lenchik
- Division of Endocrinology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Clayton Mathews
- Division of Endocrinology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Alberto Pugliese
- Division of Endocrinology, Department of Microbiology and Immunology, Department of Medicine, Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Domenica A McCarthy
- Glycation and Diabetes Complications, Mater Research Institute, Translational Research Institute (TRI), The University of Queensland (MRI-UQ), 37 Kent Street, Woolloongabba, Brisbane, QLD, 4102, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Selena Le Bagge
- Glycation and Diabetes Complications, Mater Research Institute, Translational Research Institute (TRI), The University of Queensland (MRI-UQ), 37 Kent Street, Woolloongabba, Brisbane, QLD, 4102, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Adam Ewing
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Translational Bioinformatics Group, MRI-UQ, TRI, Brisbane, Australia
| | - Mark Harris
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Queensland Diabetes Centre, Mater Health Services, Brisbane, Australia
| | - Kristen J Radford
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Cancer Immunotherapies Group, MRI-UQ, TRI, Brisbane, Australia
| | - Danielle J Borg
- Glycation and Diabetes Complications, Mater Research Institute, Translational Research Institute (TRI), The University of Queensland (MRI-UQ), 37 Kent Street, Woolloongabba, Brisbane, QLD, 4102, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Ivan Gerling
- Division of Endocrinology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Josephine M Forbes
- Glycation and Diabetes Complications, Mater Research Institute, Translational Research Institute (TRI), The University of Queensland (MRI-UQ), 37 Kent Street, Woolloongabba, Brisbane, QLD, 4102, Australia.
- Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
11
|
Brooks EP, Sussel L. Not the second fiddle: α cell development, identity, and function in health and diabetes. J Endocrinol 2023; 258:e220297. [PMID: 37171828 PMCID: PMC10524258 DOI: 10.1530/joe-22-0297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 05/12/2023] [Indexed: 05/13/2023]
Abstract
Historic and emerging studies provide evidence for the deterioration of pancreatic α cell function and identity in diabetes mellitus. Increased access to human tissue and the availability of more sophisticated molecular technologies have identified key insights into how α cell function and identity are preserved in healthy conditions and how they become dysfunctional in response to stress. These studies have revealed evidence of impaired glucagon secretion, shifts in α cell electrophysiology, changes in α cell mass, dysregulation of α cell transcription, and α-to-β cell conversion prior to and during diabetes. In this review, we outline the current state of research on α cell identity in health and disease. Evidence in model organisms and humans suggests that in addition to β cell dysfunction, diabetes is associated with a fundamental dysregulation of α cell identity. Importantly, epigenetic studies have revealed that α cells retain more poised and open chromatin at key cell-specific and diabetes-dysregulated genes, supporting the model that the inherent epigenetic plasticity of α cells makes them susceptible to the transcriptional changes that potentiate the loss of identity and function seen in diabetes. Thus, additional research into the maintenance of α cell identity and function is critical to fully understanding diabetes. Furthermore, these studies suggest α cells could represent an alternative source of new β cells for diabetes treatment.
Collapse
Affiliation(s)
- Elliott P Brooks
- Barbara Davis Center for Diabetes, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lori Sussel
- Barbara Davis Center for Diabetes, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
12
|
Xie B, Gao D, Zhou B, Chen S, Wang L. New discoveries in the field of metabolism by applying single-cell and spatial omics. J Pharm Anal 2023; 13:711-725. [PMID: 37577385 PMCID: PMC10422156 DOI: 10.1016/j.jpha.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 08/15/2023] Open
Abstract
Single-cell multi-Omics (SCM-Omics) and spatial multi-Omics (SM-Omics) technologies provide state-of-the-art methods for exploring the composition and function of cell types in tissues/organs. Since its emergence in 2009, single-cell RNA sequencing (scRNA-seq) has yielded many groundbreaking new discoveries. The combination of this method with the emergence and development of SM-Omics techniques has been a pioneering strategy in neuroscience, developmental biology, and cancer research, especially for assessing tumor heterogeneity and T-cell infiltration. In recent years, the application of these methods in the study of metabolic diseases has also increased. The emerging SCM-Omics and SM-Omics approaches allow the molecular and spatial analysis of cells to explore regulatory states and determine cell fate, and thus provide promising tools for unraveling heterogeneous metabolic processes and making them amenable to intervention. Here, we review the evolution of SCM-Omics and SM-Omics technologies, and describe the progress in the application of SCM-Omics and SM-Omics in metabolism-related diseases, including obesity, diabetes, nonalcoholic fatty liver disease (NAFLD) and cardiovascular disease (CVD). We also conclude that the application of SCM-Omics and SM-Omics approaches can help resolve the molecular mechanisms underlying the pathogenesis of metabolic diseases in the body and facilitate therapeutic measures for metabolism-related diseases. This review concludes with an overview of the current status of this emerging field and the outlook for its future.
Collapse
Affiliation(s)
- Baocai Xie
- Department of Critical Care Medicine, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, 518060, China
- Department of Respiratory Diseases, The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450014, China
| | - Dengfeng Gao
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Biqiang Zhou
- Department of Geriatric & Spinal Pain Multi-Department Treatment, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen, Guangdong, 518035, China
| | - Shi Chen
- Department of Critical Care Medicine, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, 518060, China
- Department of Gastroenterology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Lianrong Wang
- Department of Respiratory Diseases, The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450014, China
- Department of Gastroenterology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
13
|
Kawamori D, Sasaki S. Newly discovered knowledge pertaining to glucagon and its clinical applications. J Diabetes Investig 2023. [PMID: 37052948 DOI: 10.1111/jdi.14009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 03/10/2023] [Indexed: 04/14/2023] Open
Abstract
Glucagon has been defined as an 'insulin counteracting hormone', which raises blood glucose levels. Recent progress in basic research has shown that glucagon is closely involved in glucose and amino acid metabolism. Additionally, its secretion is intricately, but precisely, regulated by various mechanisms involving molecules in addition to glucose, thus showing its critical role in systemic nutrient metabolism. An innovative dual-antibody-linked immunosorbent assay for glucagon that improves measurement accuracy has been developed, and substantial clinical findings have been obtained using this new system. This discovery expanded the pathophysiological significance of glucagon and accelerated the development of its clinical applications in diabetes.
Collapse
Grants
- 21K08576 Ministry of Education, Culture, Sports, Science, and Technology in Japan
- 21K20902 Ministry of Education, Culture, Sports, Science, and Technology in Japan
- 22K16395 Ministry of Education, Culture, Sports, Science, and Technology in Japan
Collapse
Affiliation(s)
- Dan Kawamori
- Medical Education Center, Faculty of Medicine, Osaka University, Osaka, Japan
- Postgraduate Medical Training Center, Osaka University Hospital, Osaka University, Osaka, Japan
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shugo Sasaki
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
14
|
Castell AL, Goubault C, Ethier M, Fergusson G, Tremblay C, Baltz M, Dal Soglio D, Ghislain J, Poitout V. β Cell mass expansion during puberty involves serotonin signaling and determines glucose homeostasis in adulthood. JCI Insight 2022; 7:160854. [PMID: 36107617 PMCID: PMC9675460 DOI: 10.1172/jci.insight.160854] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 09/14/2022] [Indexed: 01/07/2023] Open
Abstract
Puberty is associated with transient insulin resistance that normally recedes at the end of puberty; however, in overweight children, insulin resistance persists, leading to an increased risk of type 2 diabetes. The mechanisms whereby pancreatic β cells adapt to pubertal insulin resistance, and how they are affected by the metabolic status, have not been investigated. Here, we show that puberty is associated with a transient increase in β cell proliferation in rats and humans of both sexes. In rats, β cell proliferation correlated with a rise in growth hormone (GH) levels. Serum from pubertal rats and humans promoted β cell proliferation, suggesting the implication of a circulating factor. In pubertal rat islets, expression of genes of the GH/serotonin (5-hydroxytryptamine [5-HT]) pathway underwent changes consistent with a proliferative effect. Inhibition of the pro-proliferative 5-HT receptor isoform HTR2B blocked the increase in β cell proliferation in pubertal islets ex vivo and in vivo. Peripubertal metabolic stress blunted β cell proliferation during puberty and led to altered glucose homeostasis later in life. This study identifies a role of GH/GH receptor/5-HT/HTR2B signaling in the control of β cell mass expansion during puberty and identifies a mechanistic link between pubertal obesity and the risk of developing type 2 diabetes.
Collapse
Affiliation(s)
- Anne-Laure Castell
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Medicine and
| | - Clara Goubault
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Pharmacology and Physiology, University of Montreal, Quebec, Canada
| | - Mélanie Ethier
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Grace Fergusson
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Caroline Tremblay
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Marie Baltz
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Dorothée Dal Soglio
- CHU Sainte-Justine, Montreal, Quebec, Canada.,Department of Pathology and Cell Biology, University of Montreal, Montreal, Quebec, Canada
| | - Julien Ghislain
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Medicine and
| |
Collapse
|
15
|
Tudurí E, Soriano S, Almagro L, Montanya E, Alonso-Magdalena P, Nadal Á, Quesada I. The pancreatic β-cell in ageing: Implications in age-related diabetes. Ageing Res Rev 2022; 80:101674. [PMID: 35724861 DOI: 10.1016/j.arr.2022.101674] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/07/2022] [Accepted: 06/14/2022] [Indexed: 11/15/2022]
Abstract
The prevalence of type 2 diabetes (T2D) and impaired glucose tolerance (IGT) increases with ageing. T2D generally results from progressive impairment of the pancreatic islets to adapt β-cell mass and function in the setting of insulin resistance and increased insulin demand. Several studies have shown an age-related decline in peripheral insulin sensitivity. However, a precise understanding of the pancreatic β-cell response in ageing is still lacking. In this review, we summarize the age-related alterations, adaptations and/or failures of β-cells at the molecular, morphological and functional levels in mouse and human. Age-associated alterations include processes such as β-cell proliferation, apoptosis and cell identity that can influence β-cell mass. Age-related changes also affect β-cell function at distinct steps including electrical activity, Ca2+ signaling and insulin secretion, among others. We will consider the potential impact of these alterations and those mediated by senescence pathways on β-cells and their implications in age-related T2D. Finally, given the great diversity of results in the field of β-cell ageing, we will discuss the sources of this heterogeneity. A better understanding of β-cell biology during ageing, particularly at older ages, will improve our insight into the contribution of β-cells to age-associated T2D and may boost new therapeutic strategies.
Collapse
Affiliation(s)
- Eva Tudurí
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Elche, Spain; Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain; Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain.
| | - Sergi Soriano
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Elche, Spain; Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Lucía Almagro
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Elche, Spain
| | - Eduard Montanya
- Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain; Department of Clinical Sciences, University of Barcelona, Barcelona, Spain; Bellvitge Hospital-IDIBELL, Barcelona, Spain, University of Barcelona, Barcelona, Spain
| | - Paloma Alonso-Magdalena
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Elche, Spain; Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Ángel Nadal
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Elche, Spain; Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Ivan Quesada
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Elche, Spain; Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain.
| |
Collapse
|
16
|
Dai XQ, Camunas-Soler J, Briant LJB, Dos Santos T, Spigelman AF, Walker EM, Arrojo E Drigo R, Bautista A, Jones RC, Avrahami D, Lyon J, Nie A, Smith N, Zhang Y, Johnson J, Manning Fox JE, Michelakis ED, Light PE, Kaestner KH, Kim SK, Rorsman P, Stein RW, Quake SR, MacDonald PE. Heterogenous impairment of α cell function in type 2 diabetes is linked to cell maturation state. Cell Metab 2022; 34:256-268.e5. [PMID: 35108513 PMCID: PMC8852281 DOI: 10.1016/j.cmet.2021.12.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/08/2021] [Accepted: 12/22/2021] [Indexed: 02/03/2023]
Abstract
In diabetes, glucagon secretion from pancreatic α cells is dysregulated. The underlying mechanisms, and whether dysfunction occurs uniformly among cells, remain unclear. We examined α cells from human donors and mice using electrophysiological, transcriptomic, and computational approaches. Rising glucose suppresses α cell exocytosis by reducing P/Q-type Ca2+ channel activity, and this is disrupted in type 2 diabetes (T2D). Upon high-fat feeding of mice, α cells shift toward a "β cell-like" electrophysiological profile in concert with indications of impaired identity. In human α cells we identified links between cell membrane properties and cell surface signaling receptors, mitochondrial respiratory chain complex assembly, and cell maturation. Cell-type classification using machine learning of electrophysiology data demonstrated a heterogenous loss of "electrophysiologic identity" in α cells from donors with type 2 diabetes. Indeed, a subset of α cells with impaired exocytosis is defined by an enrichment in progenitor and lineage markers and upregulation of an immature transcriptomic phenotype, suggesting important links between α cell maturation state and dysfunction.
Collapse
Affiliation(s)
- Xiao-Qing Dai
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Joan Camunas-Soler
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94518, USA
| | - Linford J B Briant
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, Churchill Hospital, Oxford OX3 7LE, UK
| | - Theodore Dos Santos
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Aliya F Spigelman
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Emily M Walker
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48105, USA
| | - Rafael Arrojo E Drigo
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Austin Bautista
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Robert C Jones
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Dana Avrahami
- Endocrinology and Metabolism Department, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - James Lyon
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Aifang Nie
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Nancy Smith
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Yongneng Zhang
- Department of Medicine, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Janyne Johnson
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Jocelyn E Manning Fox
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G2R3, Canada
| | | | - Peter E Light
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA 94305, USA
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, Churchill Hospital, Oxford OX3 7LE, UK
| | - Roland W Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94518, USA; Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Patrick E MacDonald
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G2R3, Canada.
| |
Collapse
|
17
|
Fu H, Sun H, Kong H, Lou B, Chen H, Zhou Y, Huang C, Qin L, Shan Y, Dai S. Discoveries in Pancreatic Physiology and Disease Biology Using Single-Cell RNA Sequencing. Front Cell Dev Biol 2022; 9:732776. [PMID: 35141228 PMCID: PMC8819087 DOI: 10.3389/fcell.2021.732776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 12/15/2021] [Indexed: 11/16/2022] Open
Abstract
Transcriptome analysis is used to study gene expression in human tissues. It can promote the discovery of new therapeutic targets for related diseases by characterizing the endocrine function of pancreatic physiology and pathology, as well as the gene expression of pancreatic tumors. Compared to whole-tissue RNA sequencing, single-cell RNA sequencing (scRNA-seq) can detect transcriptional activity within a single cell. The scRNA-seq had an invaluable contribution to discovering previously unknown cell subtypes in normal and diseased pancreases, studying the functional role of rare islet cells, and studying various types of cells in diabetes as well as cancer. Here, we review the recent in vitro and in vivo advances in understanding the pancreatic physiology and pathology associated with single-cell sequencing technology, which may provide new insights into treatment strategy optimization for diabetes and pancreatic cancer.
Collapse
Affiliation(s)
- Haotian Fu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongwei Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Wenzhou, China
| | - Hongru Kong
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bin Lou
- Department of Surgery, The Third People’s Hospital of Yuhang District, Hangzhou, China
| | - Hao Chen
- Department of Thyroid Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yilin Zhou
- Department of Biology, Boston University, Boston, MA, United States
| | - Chaohao Huang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lei Qin
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Lei Qin, ; Yunfeng Shan, ; Shengjie Dai,
| | - Yunfeng Shan
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Wenzhou, China
- *Correspondence: Lei Qin, ; Yunfeng Shan, ; Shengjie Dai,
| | - Shengjie Dai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Lei Qin, ; Yunfeng Shan, ; Shengjie Dai,
| |
Collapse
|
18
|
Holter MM, Saikia M, Cummings BP. Alpha-cell paracrine signaling in the regulation of beta-cell insulin secretion. Front Endocrinol (Lausanne) 2022; 13:934775. [PMID: 35957816 PMCID: PMC9360487 DOI: 10.3389/fendo.2022.934775] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/28/2022] [Indexed: 01/14/2023] Open
Abstract
As an incretin hormone, glucagon-like peptide 1 (GLP-1) lowers blood glucose levels by enhancing glucose-stimulated insulin secretion from pancreatic beta-cells. Therapies targeting the GLP-1 receptor (GLP-1R) use the classical incretin model as a physiological framework in which GLP-1 secreted from enteroendocrine L-cells acts on the beta-cell GLP-1R. However, this model has come into question, as evidence demonstrating local, intra-islet GLP-1 production has advanced the competing hypothesis that the incretin activity of GLP-1 may reflect paracrine signaling of GLP-1 from alpha-cells on GLP-1Rs on beta-cells. Additionally, recent studies suggest that alpha-cell-derived glucagon can serve as an additional, albeit less potent, ligand for the beta-cell GLP-1R, thereby expanding the role of alpha-cells beyond that of a counterregulatory cell type. Efforts to understand the role of the alpha-cell in the regulation of islet function have revealed both transcriptional and functional heterogeneity within the alpha-cell population. Further analysis of this heterogeneity suggests that functionally distinct alpha-cell subpopulations display alterations in islet hormone profile. Thus, the role of the alpha-cell in glucose homeostasis has evolved in recent years, such that alpha-cell to beta-cell communication now presents a critical axis regulating the functional capacity of beta-cells. Herein, we describe and integrate recent advances in our understanding of the impact of alpha-cell paracrine signaling on insulin secretory dynamics and how this intra-islet crosstalk more broadly contributes to whole-body glucose regulation in health and under metabolic stress. Moreover, we explore how these conceptual changes in our understanding of intra-islet GLP-1 biology may impact our understanding of the mechanisms of incretin-based therapeutics.
Collapse
Affiliation(s)
- Marlena M. Holter
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
- *Correspondence: Marlena M. Holter,
| | - Mridusmita Saikia
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Bethany P. Cummings
- School of Medicine, Department of Surgery, Center for Alimentary and Metabolic Sciences, University of California, Davis, Sacramento, CA, United States
| |
Collapse
|
19
|
Martínez MS, Manzano A, Olivar LC, Nava M, Salazar J, D’Marco L, Ortiz R, Chacín M, Guerrero-Wyss M, Cabrera de Bravo M, Cano C, Bermúdez V, Angarita L. The Role of the α Cell in the Pathogenesis of Diabetes: A World beyond the Mirror. Int J Mol Sci 2021; 22:9504. [PMID: 34502413 PMCID: PMC8431704 DOI: 10.3390/ijms22179504] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022] Open
Abstract
Type 2 Diabetes Mellitus (T2DM) is one of the most prevalent chronic metabolic disorders, and insulin has been placed at the epicentre of its pathophysiological basis. However, the involvement of impaired alpha (α) cell function has been recognized as playing an essential role in several diseases, since hyperglucagonemia has been evidenced in both Type 1 and T2DM. This phenomenon has been attributed to intra-islet defects, like modifications in pancreatic α cell mass or dysfunction in glucagon's secretion. Emerging evidence has shown that chronic hyperglycaemia provokes changes in the Langerhans' islets cytoarchitecture, including α cell hyperplasia, pancreatic beta (β) cell dedifferentiation into glucagon-positive producing cells, and loss of paracrine and endocrine regulation due to β cell mass loss. Other abnormalities like α cell insulin resistance, sensor machinery dysfunction, or paradoxical ATP-sensitive potassium channels (KATP) opening have also been linked to glucagon hypersecretion. Recent clinical trials in phases 1 or 2 have shown new molecules with glucagon-antagonist properties with considerable effectiveness and acceptable safety profiles. Glucagon-like peptide-1 (GLP-1) agonists and Dipeptidyl Peptidase-4 inhibitors (DPP-4 inhibitors) have been shown to decrease glucagon secretion in T2DM, and their possible therapeutic role in T1DM means they are attractive as an insulin-adjuvant therapy.
Collapse
Affiliation(s)
- María Sofía Martínez
- MedStar Health Internal Medicine, Georgetown University Affiliated, Baltimore, MD 21218-2829, USA;
| | - Alexander Manzano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, Universidad del Zulia, Maracaibo 4002, Venezuela; (A.M.); (L.C.O.); (M.N.); (J.S.); (C.C.)
| | - Luis Carlos Olivar
- Endocrine and Metabolic Diseases Research Center, School of Medicine, Universidad del Zulia, Maracaibo 4002, Venezuela; (A.M.); (L.C.O.); (M.N.); (J.S.); (C.C.)
| | - Manuel Nava
- Endocrine and Metabolic Diseases Research Center, School of Medicine, Universidad del Zulia, Maracaibo 4002, Venezuela; (A.M.); (L.C.O.); (M.N.); (J.S.); (C.C.)
| | - Juan Salazar
- Endocrine and Metabolic Diseases Research Center, School of Medicine, Universidad del Zulia, Maracaibo 4002, Venezuela; (A.M.); (L.C.O.); (M.N.); (J.S.); (C.C.)
| | - Luis D’Marco
- Department of Nephrology, Hospital Clinico Universitario de Valencia, INCLIVA, University of Valencia, 46010 Valencia, Spain;
| | - Rina Ortiz
- Facultad de Medicina, Universidad Católica de Cuenca, Ciudad de Cuenca, Azuay 010105, Ecuador;
| | - Maricarmen Chacín
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080022, Colombia; (M.C.); (V.B.)
| | - Marion Guerrero-Wyss
- Escuela de Nutrición y Dietética, Facultad de Ciencias Para el Cuidado de la Salud, Universidad San Sebastián, Valdivia 5090000, Chile;
| | | | - Clímaco Cano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, Universidad del Zulia, Maracaibo 4002, Venezuela; (A.M.); (L.C.O.); (M.N.); (J.S.); (C.C.)
| | - Valmore Bermúdez
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080022, Colombia; (M.C.); (V.B.)
| | - Lisse Angarita
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andres Bello, Sede Concepción 4260000, Chile
| |
Collapse
|
20
|
Oropeza D, Cigliola V, Romero A, Chera S, Rodríguez-Seguí SA, Herrera PL. Stage-specific transcriptomic changes in pancreatic α-cells after massive β-cell loss. BMC Genomics 2021; 22:585. [PMID: 34340653 PMCID: PMC8330016 DOI: 10.1186/s12864-021-07812-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 06/10/2021] [Indexed: 12/29/2022] Open
Abstract
Background Loss of pancreatic insulin-secreting β-cells due to metabolic or autoimmune damage leads to the development of diabetes. The discovery that α-cells can be efficiently reprogrammed into insulin-secreting cells in mice and humans has opened promising avenues for innovative diabetes therapies. β-cell loss triggers spontaneous reprogramming of only 1–2% of α-cells, limiting the extent of regeneration. Most α-cells are refractory to conversion and their global transcriptomic response to severe β-cell loss as well as the mechanisms opposing their reprogramming into insulin producers are largely unknown. Here, we performed RNA-seq on FAC-sorted α-cells to characterize their global transcriptional responses at different time points after massive β-cell ablation. Results Our results show that α-cells undergo stage-specific transcriptional changes 5- and 15-days post-diphtheria toxin (DT)-mediated β-cell ablation. At 5 days, α-cells transiently upregulate various genes associated with interferon signaling and proliferation, including Interferon Induced Protein with Tetratricopeptide Repeats 3 (Ifit3). Subsequently, at 15 days post β-cell ablation, α-cells undergo a transient downregulation of genes from several pathways including Insulin receptor, mTOR and MET signaling. Conclusions The results presented here pinpoint novel markers discriminating α-cells at different stages after acute β-cell loss, and highlight additional signaling pathways that are modulated in α-cells in this context. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07812-x.
Collapse
Affiliation(s)
- Daniel Oropeza
- Department of Genetic Medicine and Development, iGE3 and Centre Facultaire du Diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Valentina Cigliola
- Department of Genetic Medicine and Development, iGE3 and Centre Facultaire du Diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Present address: Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.,Regeneration Next, Duke University, Durham, North Carolina, 27710, USA
| | - Agustín Romero
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET-Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina
| | - Simona Chera
- Department of Genetic Medicine and Development, iGE3 and Centre Facultaire du Diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Clinical Science, Center for Diabetes Research, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Santiago A Rodríguez-Seguí
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET-Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina.
| | - Pedro L Herrera
- Department of Genetic Medicine and Development, iGE3 and Centre Facultaire du Diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
21
|
GLP-1 peptide analogs for targeting pancreatic beta cells. Drug Discov Today 2021; 26:1936-1943. [PMID: 33839290 DOI: 10.1016/j.drudis.2021.03.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/01/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023]
Abstract
Loss or dysfunction of the pancreatic beta cells or insulin receptors leads to diabetes mellitus (DM). This usually occurs over many years; therefore, the development of methods for the timely detection and clinical intervention are vital to prevent the development of this disease. Glucagon-like peptide-1 receptor (GLP-1R) is the receptor of GLP-1, an incretin hormone that causes insulin secretion in a glucose-dependent manner. GLP-1R is highly expressed on the surface of pancreatic beta cells, providing a potential target for bioimaging. In this review, we provide an overview of various strategies, such as the development of GLP-1R agonists (e.g., exendin-4), and GLP-1 sequence modifications for GLP-1R targeting for the diagnosis and treatment of pancreatic beta cell disorders. We also discuss the challenges of targeting pancreatic beta cells and strategies to address such challenges.
Collapse
|
22
|
Seiron P, Stenwall A, Hedin A, Granlund L, Esguerra JLS, Volkov P, Renström E, Korsgren O, Lundberg M, Skog O. Transcriptional analysis of islets of Langerhans from organ donors of different ages. PLoS One 2021; 16:e0247888. [PMID: 33711030 PMCID: PMC7954335 DOI: 10.1371/journal.pone.0247888] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
Insulin secretion is impaired with increasing age. In this study, we aimed to determine whether aging induces specific transcriptional changes in human islets. Laser capture microdissection was used to extract pancreatic islet tissue from 37 deceased organ donors aged 1-81 years. The transcriptomes of the extracted islets were analysed using Ion AmpliSeq sequencing. 346 genes that co-vary significantly with age were found. There was an increased transcription of genes linked to senescence, and several aspects of the cell cycle machinery were downregulated with increasing age. We detected numerous genes not linked to aging in previous studies likely because earlier studies analysed islet cells isolated by enzymatic digestion which might affect the islet transcriptome. Among the novel genes demonstrated to correlate with age, we found an upregulation of SPP1 encoding osteopontin. In beta cells, osteopontin has been seen to be protective against both cytotoxicity and hyperglycaemia. In summary, we present a transcriptional profile of aging in human islets and identify genes that could affect disease course in diabetes.
Collapse
Affiliation(s)
- Peter Seiron
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Anton Stenwall
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Anders Hedin
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Louise Granlund
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | | - Petr Volkov
- Department of Clinical Sciences-Malmö, Lund University Diabetes Centre, Malmö, Sweden
| | - Erik Renström
- Department of Clinical Sciences-Malmö, Lund University Diabetes Centre, Malmö, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Lundberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Oskar Skog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
23
|
Abdelalim EM. Modeling different types of diabetes using human pluripotent stem cells. Cell Mol Life Sci 2021; 78:2459-2483. [PMID: 33242105 PMCID: PMC11072720 DOI: 10.1007/s00018-020-03710-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/19/2020] [Accepted: 11/11/2020] [Indexed: 12/22/2022]
Abstract
Diabetes mellitus (DM) is a metabolic disease characterized by chronic hyperglycemia as a result of progressive loss of pancreatic β cells, which could lead to several debilitating complications. Different paths, triggered by several genetic and environmental factors, lead to the loss of pancreatic β cells and/or function. Understanding these many paths to β cell damage or dysfunction could help in identifying therapeutic approaches specific for each path. Most of our knowledge about diabetes pathophysiology has been obtained from studies on animal models, which do not fully recapitulate human diabetes phenotypes. Currently, human pluripotent stem cell (hPSC) technology is a powerful tool for generating in vitro human models, which could provide key information about the disease pathogenesis and provide cells for personalized therapies. The recent progress in generating functional hPSC-derived β cells in combination with the rapid development in genomic and genome-editing technologies offer multiple options to understand the cellular and molecular mechanisms underlying the development of different types of diabetes. Recently, several in vitro hPSC-based strategies have been used for studying monogenic and polygenic forms of diabetes. This review summarizes the current knowledge about different hPSC-based diabetes models and how these models improved our current understanding of the pathophysiology of distinct forms of diabetes. Also, it highlights the progress in generating functional β cells in vitro, and discusses the current challenges and future perspectives related to the use of the in vitro hPSC-based strategies.
Collapse
Affiliation(s)
- Essam M Abdelalim
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar.
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha, Qatar.
| |
Collapse
|
24
|
Oakie A, Nostro MC. Harnessing Proliferation for the Expansion of Stem Cell-Derived Pancreatic Cells: Advantages and Limitations. Front Endocrinol (Lausanne) 2021; 12:636182. [PMID: 33716986 PMCID: PMC7947602 DOI: 10.3389/fendo.2021.636182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/20/2021] [Indexed: 12/13/2022] Open
Abstract
Restoring the number of glucose-responsive β-cells in patients living with diabetes is critical for achieving normoglycemia since functional β-cells are lost during the progression of both type 1 and 2 diabetes. Stem cell-derived β-cell replacement therapies offer an unprecedented opportunity to replace the lost β-cell mass, yet differentiation efficiencies and the final yield of insulin-expressing β-like cells are low when using established protocols. Driving cellular proliferation at targeted points during stem cell-derived pancreatic progenitor to β-like cell differentiation can serve as unique means to expand the final cell therapeutic product needed to restore insulin levels. Numerous studies have examined the effects of β-cell replication upon functionality, using primary islets in vitro and mouse models in vivo, yet studies that focus on proliferation in stem cell-derived pancreatic models are only just emerging in the field. This mini review will discuss the current literature on cell proliferation in pancreatic cells, with a focus on the proliferative state of stem cell-derived pancreatic progenitors and β-like cells during their differentiation and maturation. The benefits of inducing proliferation to increase the final number of β-like cells will be compared against limitations associated with driving replication, such as the blunted capacity of proliferating β-like cells to maintain optimal β-cell function. Potential strategies that may bypass the challenges induced by the up-regulation of cell cycle-associated factors during β-cell differentiation will be proposed.
Collapse
Affiliation(s)
- Amanda Oakie
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Maria Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
25
|
Shi J, Zhang YQ, Hao DD, Fu SH, Meng JL. Key regulatory genes and signaling pathways involved in islet culture: a bioinformatic analysis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2021; 14:292-303. [PMID: 33564361 PMCID: PMC7868784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/31/2020] [Indexed: 06/12/2023]
Abstract
Type 1 diabetes (T1D) is characterized by non-ideal mass and low survival rate of islets. Therefore, it is necessary to find intrinsic factors that prolong the survival of islets. This study aimed to track out hub genes and pathways in the process of islet culture by bioinformatic analysis. We downloaded the gene expression microarray of GSE42591 from the Gene Expression Omnibus (GEO). Aberrant Differentially methylated genes (DMGs) were obtained using the GEO2R tool. Gene ontology (GO) analysis and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway enrichment analyses were performed on selected genes by using the Database for Annotation Visualization and Integrated Discovery (DAVID). A protein-protein interaction (PPI) network was constructed with the Retrieval of Interacting Genes (STRING) and visualized in Cytoscape 3.7.2. A total of 434 genes were overexpressed and 114 genes underexpressed in fresh to cultured 4 h tissue. KEGG pathway enrichment analyses revealed the TGF-beta signaling pathway, MAPK signaling pathway, or VEGF signaling pathway. The genes FN1, MKI67, IGF1, MAPK14, COL1A1 might be involved in islet culture. In general, this work scrutinized islet culture-relevant knowledge and provided insight into the regulation and mediation of islet survival.
Collapse
Affiliation(s)
- Jing Shi
- Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region (Hospital. C.T.)No. 20, Xi Mian Qiao Heng Jie, Wuhou District, Chengdu 610041, Sichuan Province, China
| | - Yong-Qun Zhang
- Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region (Hospital. C.T.)No. 20, Xi Mian Qiao Heng Jie, Wuhou District, Chengdu 610041, Sichuan Province, China
| | - Dou-Dou Hao
- Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region (Hospital. C.T.)No. 20, Xi Mian Qiao Heng Jie, Wuhou District, Chengdu 610041, Sichuan Province, China
| | - Su-Hong Fu
- Lab of Natural Medicine of West China Hospital of West China Medical School of Sichuan UniversityNo. 88, South Keyuan Road, Chengdu High-Tech Zone, Chengdu 610041, Sichuan Province, China
| | - Jin-Li Meng
- Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region (Hospital. C.T.)No. 20, Xi Mian Qiao Heng Jie, Wuhou District, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
26
|
Cong GZ, Ghosh KK, Mishra S, Gulyás M, Kovács T, Máthé D, Padmanabhan P, Gulyás B. Targeted pancreatic beta cell imaging for early diagnosis. Eur J Cell Biol 2020; 99:151110. [PMID: 33070042 DOI: 10.1016/j.ejcb.2020.151110] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 06/29/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic beta cells are important in blood glucose level regulation. As type 1 and 2 diabetes are getting prevalent worldwide, we need to explore new methods for early detection of beta cell-related afflictions. Using bioimaging techniques to measure beta cell mass is crucial because a decrease in beta cell density is seen in diseases such as diabetes and thus can be a new way of diagnosis for such diseases. We also need to appraise beta cell purity in transplanted islets for type 1 diabetes patients. Sufficient amount of functional beta cells must also be determined before being transplanted to the patients. In this review, indirect imaging of beta cells will be discussed. This includes membrane protein on pancreatic beta cells whereby specific probes are designed for different imaging modalities mainly magnetic resonance imaging, positron emission tomography and fluorescence imaging. Direct imaging of insulin is also explored though probes synthesized for such function are relatively fewer. The path for successful pancreatic beta cell imaging is fraught with challenges like non-specific binding, lack of beta cell-restricted targets, the requirement of probes to cross multiple lipid layers to bind to intracellular insulin. Hence, there is an urgent need to develop new imaging techniques and innovative probing constructs in the entire imaging chain of bioengineering to provide early detection of beta cell-related pathology.
Collapse
Affiliation(s)
- Goh Zheng Cong
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Krishna Kanta Ghosh
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Sachin Mishra
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Miklós Gulyás
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskölds väg 20, Uppsala Se-751 85, Sweden
| | - Tibor Kovács
- Institute of Radiochemistry and Radioecology, University of Pannonia, Egyetem u. 10, H-8200 Veszprém, Hungary
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology, Semmelweis University Faculty of Medicine, Tűzoltó u. 37-47, Budapest H-1094, Hungary
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore.
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore.
| |
Collapse
|
27
|
Horton TM, Sundaram V, Lee CHJ, Hornbacker K, Van Vleck A, Benjamin KN, Zemek A, Longacre TA, Kunz PL, Annes JP. PAM staining intensity of primary neuroendocrine neoplasms is a potential prognostic biomarker. Sci Rep 2020; 10:10943. [PMID: 32616904 PMCID: PMC7331689 DOI: 10.1038/s41598-020-68071-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
Neuroendocrine neoplasms (NENs) are rare epithelial tumors with heterogeneous and frequently unpredictable clinical behavior. Available biomarkers are insufficient to guide individual patient prognosis or therapy selection. Peptidylglycine α-amidating monooxygenase (PAM) is an enzyme expressed by neuroendocrine cells that participates in hormone maturation. The objective of this study was to assess the distribution, clinical associations and survival implications of PAM immunoreactivity in primary NENs. Of 109 primary NENs, 7% were PAM-negative, 25% were PAM-low and 68% were PAM-high. Staining intensity was high in small bowel (p = 0.04) and low in stomach (p = 0.004) NENs. PAM staining was lower in higher grade tumors (p < 0.001) and patients who died (p < 0.001) but did not vary by tumor size or stage at surgery. In patients who died, time to death was shorter in patients with reduced PAM immunoreactivity: median times to death were 11.3 (PAM-negative), 29.4 (PAM-low) and 61.7 (PAM-high) months. Lower PAM staining was associated with increased risk of death after adjusting for disease stage [PAM negative, HR = 13.8 (CI: 4.2–45.5)]. PAM immunoreactivity in primary NENs is readily assessable and a potentially useful stage-independent predictor of survival.
Collapse
Affiliation(s)
- Timothy M Horton
- Department of Chemistry, Stanford University, Stanford, CA, USA.,Chemistry, Engineering and Medicine for Human Health (ChEM-H) Institute, Stanford University, Stanford, CA, USA
| | - Vandana Sundaram
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Christine Hye-Jin Lee
- Division of Endocrinology, Department of Medicine, Stanford University, CCSR 2255-A, 1291 Welch Rd., Stanford, CA, 94305-5165, USA
| | - Kathleen Hornbacker
- Endocrine Oncology Program, Stanford University, Stanford, USA.,Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Aidan Van Vleck
- Division of Endocrinology, Department of Medicine, Stanford University, CCSR 2255-A, 1291 Welch Rd., Stanford, CA, 94305-5165, USA
| | - Kaisha N Benjamin
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Allison Zemek
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Teri A Longacre
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Pamela L Kunz
- Endocrine Oncology Program, Stanford University, Stanford, USA.,Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Justin P Annes
- Chemistry, Engineering and Medicine for Human Health (ChEM-H) Institute, Stanford University, Stanford, CA, USA. .,Division of Endocrinology, Department of Medicine, Stanford University, CCSR 2255-A, 1291 Welch Rd., Stanford, CA, 94305-5165, USA. .,Endocrine Oncology Program, Stanford University, Stanford, USA.
| |
Collapse
|
28
|
Gilon P. The Role of α-Cells in Islet Function and Glucose Homeostasis in Health and Type 2 Diabetes. J Mol Biol 2020; 432:1367-1394. [PMID: 31954131 DOI: 10.1016/j.jmb.2020.01.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/23/2019] [Accepted: 01/06/2020] [Indexed: 01/09/2023]
Abstract
Pancreatic α-cells are the major source of glucagon, a hormone that counteracts the hypoglycemic action of insulin and strongly contributes to the correction of acute hypoglycemia. The mechanisms by which glucose controls glucagon secretion are hotly debated, and it is still unclear to what extent this control results from a direct action of glucose on α-cells or is indirectly mediated by β- and/or δ-cells. Besides its hyperglycemic action, glucagon has many other effects, in particular on lipid and amino acid metabolism. Counterintuitively, glucagon seems also required for an optimal insulin secretion in response to glucose by acting on its cognate receptor and, even more importantly, on GLP-1 receptors. Patients with diabetes mellitus display two main alterations of glucagon secretion: a relative hyperglucagonemia that aggravates hyperglycemia, and an impaired glucagon response to hypoglycemia. Under metabolic stress states, such as diabetes, pancreatic α-cells also secrete GLP-1, a glucose-lowering hormone, whereas the gut can produce glucagon. The contribution of extrapancreatic glucagon to the abnormal glucose homeostasis is unclear. Here, I review the possible mechanisms of control of glucagon secretion and the role of α-cells on islet function in healthy state. I discuss the possible causes of the abnormal glucagonemia in diabetes, with particular emphasis on type 2 diabetes, and I briefly comment the current antidiabetic therapies affecting α-cells.
Collapse
Affiliation(s)
- Patrick Gilon
- Université Catholique de Louvain, Institute of Experimental and Clinical Research, Pole of Endocrinology, Diabetes and Nutrition, Avenue Hippocrate 55 (B1.55.06), Brussels, B-1200, Belgium.
| |
Collapse
|
29
|
Allegretti PA, Horton TM, Abdolazimi Y, Moeller HP, Yeh B, Caffet M, Michel G, Smith M, Annes JP. Generation of highly potent DYRK1A-dependent inducers of human β-Cell replication via Multi-Dimensional compound optimization. Bioorg Med Chem 2020; 28:115193. [PMID: 31757680 PMCID: PMC6941846 DOI: 10.1016/j.bmc.2019.115193] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/20/2019] [Accepted: 10/29/2019] [Indexed: 02/07/2023]
Abstract
Small molecule stimulation of β-cell regeneration has emerged as a promising therapeutic strategy for diabetes. Although chemical inhibition of dual specificity tyrosine-phosphorylation-regulated kinase 1A (DYRK1A) is sufficient to enhance β-cell replication, current lead compounds have inadequate cellular potency for in vivo application. Herein, we report the clinical stage anti-cancer kinase inhibitor OTS167 as a structurally novel, remarkably potent DYRK1A inhibitor and inducer of human β-cell replication. Unfortunately, OTS167's target promiscuity and cytotoxicity curtails utility. To tailor kinase selectivity towards DYRK1A and reduce cytotoxicity we designed a library of fifty-one OTS167 derivatives based upon a modeled structure of the DYRK1A-OTS167 complex. Indeed, derivative characterization yielded several leads with exceptional DYRK1A inhibition and human β-cell replication promoting potencies but substantially reduced cytotoxicity. These compounds are the most potent human β-cell replication-promoting compounds yet described and exemplify the potential to purposefully leverage off-target activities of advanced stage compounds for a desired application.
Collapse
Affiliation(s)
- Paul A Allegretti
- Department of Medicine and Division of Endocrinology, Stanford University, Stanford, CA 94305, USA; Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Timothy M Horton
- Department of Medicine and Division of Endocrinology, Stanford University, Stanford, CA 94305, USA; Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA; Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Yassan Abdolazimi
- Department of Medicine and Division of Endocrinology, Stanford University, Stanford, CA 94305, USA
| | - Hannah P Moeller
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA; Department of Medicine and Division of Endocrinology, Stanford University, Stanford, CA 94305, USA
| | - Benjamin Yeh
- Department of Medicine and Division of Endocrinology, Stanford University, Stanford, CA 94305, USA
| | - Matthew Caffet
- Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Guillermina Michel
- Department of Medicine and Division of Endocrinology, Stanford University, Stanford, CA 94305, USA
| | - Mark Smith
- Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Justin P Annes
- Department of Medicine and Division of Endocrinology, Stanford University, Stanford, CA 94305, USA; Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
30
|
Bru-Tari E, Oropeza D, Herrera PL. Cell Heterogeneity and Paracrine Interactions in Human Islet Function: A Perspective Focused in β-Cell Regeneration Strategies. Front Endocrinol (Lausanne) 2020; 11:619150. [PMID: 33613453 PMCID: PMC7888438 DOI: 10.3389/fendo.2020.619150] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/14/2020] [Indexed: 12/27/2022] Open
Abstract
The β-cell regeneration field has shown a strong knowledge boost in the last 10 years. Pluripotent stem cell differentiation and direct reprogramming from other adult cell types are becoming more tangible long-term diabetes therapies. Newly generated β-like-cells consistently show hallmarks of native β-cells and can restore normoglycemia in diabetic mice in virtually all recent studies. Nonetheless, these cells still show important compromises in insulin secretion, cell metabolism, electrical activity, and overall survival, perhaps due to a lack of signal integration from other islet cells. Mounting data suggest that diabetes is not only a β-cell disease, as the other islet cell types also contribute to its physiopathology. Here, we present an update on the most recent studies of islet cell heterogeneity and paracrine interactions in the context of restoring an integrated islet function to improve β-cell replacement therapies.
Collapse
|
31
|
Ahn SH, Granger A, Rankin MM, Lam CJ, Cox AR, Kushner JA. Tamoxifen suppresses pancreatic β-cell proliferation in mice. PLoS One 2019; 14:e0214829. [PMID: 31490929 PMCID: PMC6731016 DOI: 10.1371/journal.pone.0214829] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/11/2019] [Indexed: 12/13/2022] Open
Abstract
Tamoxifen is a mixed agonist/antagonist estrogen analogue that is frequently used to induce conditional gene deletion in mice using Cre-loxP mediated gene recombination. Tamoxifen is routinely employed in extremely high-doses relative to typical human doses to induce efficient gene deletion in mice. Although tamoxifen has been widely assumed to have no influence upon β-cells, the acute developmental and functional consequences of high-dose tamoxifen upon glucose homeostasis and adult β-cells are largely unknown. We tested if tamoxifen influences glucose homeostasis in male mice of various genetic backgrounds. We then carried out detailed histomorphometry studies of mouse pancreata. We also performed gene expression studies with islets of tamoxifen-treated mice and controls. Tamoxifen had modest effects upon glucose homeostasis of mixed genetic background (F1 B6129SF1/J) mice, with fasting hyperglycemia and improved glucose tolerance but without overt effects on fed glucose levels or insulin sensitivity. Tamoxifen inhibited proliferation of β-cells in a dose-dependent manner, with dramatic reductions in β-cell turnover at the highest dose (decreased by 66%). In sharp contrast, tamoxifen did not reduce proliferation of pancreatic acinar cells. β-cell proliferation was unchanged by tamoxifen in 129S2 mice but was reduced in C57Bl6 genetic background mice (decreased by 59%). Gene expression studies revealed suppression of RNA for cyclins D1 and D2 within islets of tamoxifen-treated mice. Tamoxifen has a cytostatic effect on β-cells, independent of changes in glucose homeostasis, in mixed genetic background and also in C57Bl6 mice. Tamoxifen should be used judiciously to inducibly inactivate genes in studies of glucose homeostasis.
Collapse
Affiliation(s)
- Surl-Hee Ahn
- Pediatric Endocrinology and Diabetes, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Anne Granger
- Pediatric Endocrinology and Diabetes, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Matthew M. Rankin
- Pediatric Endocrinology and Diabetes, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Carol J. Lam
- Pediatric Endocrinology and Diabetes, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, United States of America
| | - Aaron R. Cox
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, United States of America
| | - Jake A. Kushner
- Pediatric Endocrinology and Diabetes, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, United States of America
- * E-mail:
| |
Collapse
|
32
|
Bru-Tari E, Cobo-Vuilleumier N, Alonso-Magdalena P, Dos Santos RS, Marroqui L, Nadal A, Gauthier BR, Quesada I. Pancreatic alpha-cell mass in the early-onset and advanced stage of a mouse model of experimental autoimmune diabetes. Sci Rep 2019; 9:9515. [PMID: 31266981 PMCID: PMC6606577 DOI: 10.1038/s41598-019-45853-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 06/14/2019] [Indexed: 02/07/2023] Open
Abstract
Most studies in type 1 diabetes (T1D) have focused on the loss of the pancreatic beta-cell population. However, despite the involvement of the alpha-cell in the aetiology and complications of T1D, little is known about the regulation of the pancreatic alpha-cell mass in this disease. The need for a better understanding of this process is further emphasized by recent findings suggesting that alpha-cells may constitute a potential reservoir for beta-cell regeneration. In this study, we characterized the pancreatic alpha-cell mass and its regulatory processes in the transgenic RIP-B7.1 mice model of experimental autoimmune diabetes (EAD). Diabetic mice presented insulitis, hyperglycaemia, hypoinsulinemia and hyperglucagonemia along with lower pancreatic insulin content. While alpha-cell mass and pancreatic glucagon content were preserved at the early-onset of EAD, both parameters were reduced in the advanced phase. At both stages, alpha-cell size, proliferation and ductal neogenesis were up-regulated, whereas apoptosis was almost negligible. Interestingly, we found an increase in the proportion of glucagon-containing cells positive for insulin or the beta-cell transcription factor PDX1. Our findings suggest that pancreatic alpha-cell renewal mechanisms are boosted during the natural course of EAD, possibly as an attempt to maintain the alpha-cell population and/or to increase beta-cell regeneration via alpha-cell transdifferentiation.
Collapse
Affiliation(s)
- Eva Bru-Tari
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), IBMC, Universidad Miguel Hernández, Elche, Spain
- Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Nadia Cobo-Vuilleumier
- Department of Cell Regeneration and Advanced Therapies, Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain
| | - Paloma Alonso-Magdalena
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), IBMC, Universidad Miguel Hernández, Elche, Spain
- Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Reinaldo S Dos Santos
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), IBMC, Universidad Miguel Hernández, Elche, Spain
- Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Laura Marroqui
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), IBMC, Universidad Miguel Hernández, Elche, Spain
- Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Angel Nadal
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), IBMC, Universidad Miguel Hernández, Elche, Spain
- Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Benoit R Gauthier
- Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
- Department of Cell Regeneration and Advanced Therapies, Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain
| | - Ivan Quesada
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), IBMC, Universidad Miguel Hernández, Elche, Spain.
- Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain.
| |
Collapse
|
33
|
Mawla AM, Huising MO. Navigating the Depths and Avoiding the Shallows of Pancreatic Islet Cell Transcriptomes. Diabetes 2019; 68:1380-1393. [PMID: 31221802 PMCID: PMC6609986 DOI: 10.2337/dbi18-0019] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 04/29/2019] [Indexed: 12/24/2022]
Abstract
Islet gene expression has been widely studied to better understand the transcriptional features that define a healthy β-cell. Transcriptomes of FACS-purified α-, β-, and δ-cells using bulk RNA-sequencing have facilitated our understanding of the complex network of cross talk between islet cells and its effects on β-cell function. However, these approaches were by design not intended to resolve heterogeneity between individual cells. Several recent studies used single-cell RNA sequencing (scRNA-Seq) to report considerable heterogeneity within mouse and human β-cells. In this Perspective, we assess how this newfound ability to assess gene expression at single-cell resolution has enhanced our understanding of β-cell heterogeneity. We conduct a comprehensive assessment of several single human β-cell transcriptome data sets and ask if the heterogeneity reported by these studies showed overlap and concurred with previously known examples of β-cell heterogeneity. We also illustrate the impact of the inevitable limitations of working at or below the limit of detection of gene expression at single cell resolution and their consequences for the quality of single-islet cell transcriptome data. Finally, we offer some guidance on when to opt for scRNA-Seq and when bulk sequencing approaches may be better suited.
Collapse
Affiliation(s)
- Alex M Mawla
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, Davis, CA
| | - Mark O Huising
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, Davis, CA
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA
| |
Collapse
|
34
|
Lam CJ, Rankin MM, King KB, Wang MC, Shook BC, Kushner JA. Glucagon Receptor Antagonist-Stimulated α-Cell Proliferation Is Severely Restricted With Advanced Age. Diabetes 2019; 68:963-974. [PMID: 30833466 PMCID: PMC6477910 DOI: 10.2337/db18-1293] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/18/2019] [Indexed: 01/26/2023]
Abstract
Glucagon-containing α-cells potently regulate glucose homeostasis, but the developmental biology of α-cells in adults remains poorly understood. Although glucagon receptor antagonists (GRAs) have great potential as antidiabetic therapies, murine and human studies have raised concerns that GRAs might cause uncontrolled α-cell growth. Surprisingly, previous rodent GRA studies were only performed in young mice, implying that the potential impact of GRAs to drive α-cell expansion in adult patients is unclear. We assessed adaptive α-cell turnover and adaptive proliferation, administering a novel GRA (JNJ-46207382) to both young and aged mice. Basal α-cell proliferation rapidly declined soon after birth and continued to drop to very low levels in aged mice. GRA drove a 2.4-fold increase in α-cell proliferation in young mice. In contrast, GRA-induced α-cell proliferation was severely reduced in aged mice, although still present at 3.2-fold the very low basal rate of aged controls. To interrogate the lineage of GRA-induced α-cells, we sequentially administered thymidine analogs and quantified their incorporation into α-cells. Similar to previous studies of β-cells, α-cells only divided once in both basal and stimulated conditions. Lack of contribution from highly proliferative "transit-amplifying" cells supports a model whereby α-cells expand by self-renewal and not via specialized progenitors.
Collapse
Affiliation(s)
- Carol J Lam
- McNair Medical Institute, Pediatric Diabetes and Endocrinology, Baylor College of Medicine, Houston, TX
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Matthew M Rankin
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA
- Janssen Research and Development, Johnson & Johnson, Springhouse, PA
| | - Kourtney B King
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Melinda C Wang
- McNair Medical Institute, Pediatric Diabetes and Endocrinology, Baylor College of Medicine, Houston, TX
| | - Brian C Shook
- Janssen Research and Development, Johnson & Johnson, Springhouse, PA
| | - Jake A Kushner
- McNair Medical Institute, Pediatric Diabetes and Endocrinology, Baylor College of Medicine, Houston, TX
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA
| |
Collapse
|
35
|
Lam CJ, Chatterjee A, Shen E, Cox AR, Kushner JA. Low-Level Insulin Content Within Abundant Non-β Islet Endocrine Cells in Long-standing Type 1 Diabetes. Diabetes 2019; 68:598-608. [PMID: 30552110 DOI: 10.2337/db18-0305] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 11/27/2018] [Indexed: 11/13/2022]
Abstract
Although most patients with type 1 diabetes (T1D) continue to produce small amounts of insulin decades after disease onset, very few β-cells persist within their pancreata. Consequently, the source of persistent insulin secretion within T1D remains unclear. We hypothesized that low-level insulin content within non-β-cells could underlie persistent T1D insulin secretion. We tested for low levels of insulin (insulinlow) within a large cohort of JDRF Network for Pancreatic Organ Donors With Diabetes (nPOD) human pancreata across a wide range of ages and T1D disease durations. Long exposures, high-throughput imaging, and blinded parallel examiners allowed precise quantification of insulinlow cells. Of note, abundant islet endocrine cells with low quantities of insulin were present in most T1D pancreata. Insulinlow islet abundance and composition were not influenced by age, duration of diabetes, or age of onset. Insulinlow islets also contained β-cell markers at variable levels, including Pdx1, Nkx6.1, GLUT1, and PC1/3. Most insulinlow cells contained abundant glucagon and other α-cell markers, suggesting that α-cells drive much of the insulinlow phenotype in T1D. However, pancreatic polypeptide, somatostatin, and ghrelin cells also contributed to the insulinlow cell population. Insulinlow cells represent a potential source of persistent insulin secretion in long-standing T1D and a possible target for regenerative therapies to expand β-cell function in disease.
Collapse
Affiliation(s)
- Carol J Lam
- McNair Medical Institute, Baylor College of Medicine, Houston, TX
| | | | - Emily Shen
- McNair Medical Institute, Baylor College of Medicine, Houston, TX
| | - Aaron R Cox
- McNair Medical Institute, Baylor College of Medicine, Houston, TX
| | - Jake A Kushner
- McNair Medical Institute, Baylor College of Medicine, Houston, TX
| |
Collapse
|
36
|
Kawamori D. Alpha the versatile: Guardians of the islets. J Diabetes Investig 2019; 10:26-28. [PMID: 29906335 PMCID: PMC6319495 DOI: 10.1111/jdi.12875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 12/31/2022] Open
Abstract
Recent progress in α-cell research has newly revealed its versatility including secretion of multiple hormones, trans-differentiation, and abundant proliferation. A recent report from the Kushner laboratory further provided direct evidence of active proliferation of α-cells in both T1D and non-diabetic control donors. α-cells are suggested to maintain both quality and quantity of islets by serving as 'guardians' of the islets.
Collapse
Affiliation(s)
- Dan Kawamori
- Department of Metabolic MedicineGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| |
Collapse
|
37
|
Dominguez Gutierrez G, Xin Y, Okamoto H, Kim J, Lee AH, Ni M, Adler C, Yancopoulos GD, Murphy AJ, Gromada J. Gene Signature of Proliferating Human Pancreatic α Cells. Endocrinology 2018; 159:3177-3186. [PMID: 30010845 DOI: 10.1210/en.2018-00469] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/04/2018] [Indexed: 12/12/2022]
Abstract
Pancreatic α cells proliferate at a low rate, and little is known about the control of this process. Here we report the characterization of human α cells by large-scale, single-cell RNA sequencing coupled with pseudotime ordering. We identified two large subpopulations and a smaller cluster of proliferating α cells with increased expression of genes involved in cell-cycle regulation. The proliferating α cells were differentiated, had normal levels of GCG expression, and showed no signs of cellular stress. Proliferating α cells were detected in both the G1S and G2M phases of the cell cycle. Human α cells proliferate at a fivefold higher rate than human β cells and express lower levels of the cell-cycle inhibitors CDKN1A and CDKN1C. Collectively, this study provides the gene signatures of human α cells and the genes involved in their cell division. The lower expression of two cell-cycle inhibitors in human α cells could account for their higher rate of proliferation compared with their insulin-producing counterparts.
Collapse
Affiliation(s)
| | - Yurong Xin
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | - Jinrang Kim
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Ann-Hwee Lee
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Min Ni
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | | | | | | |
Collapse
|
38
|
Arany EJ, Waseem M, Strutt BJ, Chamson-Reig A, Bernardo A, Eng E, Hill DJ. Direct comparison of the abilities of bone marrow mesenchymal versus hematopoietic stem cells to reverse hyperglycemia in diabetic NOD.SCID mice. Islets 2018; 10:137-150. [PMID: 30110202 PMCID: PMC6281365 DOI: 10.1080/19382014.2018.1480285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Both bone marrow-derived hematopoietic stem cells (HSC) and mesenchymal stem cells (MSC) improve glycemic control in diabetic mice, but their kinetics and associated changes in pancreatic morphology have not been directly compared. Our goal was to examine the time course of improvements in glucose tolerance and associated changes in β-cell mass and proliferation following transplantation of equivalent numbers of HSC or MSC from the same bone marrow into diabetic non-obese diabetic severe combined immune deficiency (NOD.SCID) mice. We used transgenic mice with a targeted expression of yellow fluorescent protein (YFP) driven by the Vav1 gene promoter to genetically tag HSC and progeny. HSC were separated from bone marrow by fluorescence-activated cell sorting and MSC following cell culture. Equivalent numbers of isolated HSC or MSC were transplanted directly into the pancreas of NOD.SCID mice previously made diabetic with streptozotocin. Glucose tolerance, serum insulin, β-cell mass and β-cell proliferation were examined up to 28 days following transplant. Transplantation with MSC improved glucose tolerance within 7 days and serum insulin levels increased, but with no increase in β-cell mass. Mice transplanted with HSC showed improved glucose tolerance only after 3 weeks associated with increased β-cell proliferation and mass. We conclude that single injections of either MSC or HSC transiently improved glycemic control in diabetic NOD.SCID mice, but with different time courses. However, only HSC infiltrated the islets and were associated with an expanded β-cell mass. This suggests that MSC and HSC have differing mechanisms of action.
Collapse
Affiliation(s)
- Edith J. Arany
- Lawson Health Research Institute, London, ON, Canada
- Department of Medicine, Western University, London, ON, Canada
- Department of Pathology, Western University, London, ON, Canada
- CONTACT Dr. David J. Hill Lawson Health Research Institute, St. Joseph’s Health Care, 268 Grosvenor St, London ON Canada N6A 4V2
| | - Muhammad Waseem
- Lawson Health Research Institute, London, ON, Canada
- International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | | | | | - Adam Bernardo
- Lawson Health Research Institute, London, ON, Canada
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Elizabeth Eng
- Lawson Health Research Institute, London, ON, Canada
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - David J. Hill
- Lawson Health Research Institute, London, ON, Canada
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Department of Medicine, Western University, London, ON, Canada
| |
Collapse
|