1
|
Kamal MM, Ammar RA, Kassem DH. Silencing of forkhead box protein O-1 (FOXO-1) enhances insulin-producing cell generation from adipose mesenchymal stem cells for diabetes therapy. Life Sci 2024; 344:122579. [PMID: 38518842 DOI: 10.1016/j.lfs.2024.122579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/09/2024] [Accepted: 03/18/2024] [Indexed: 03/24/2024]
Abstract
AIMS Generation of mature β-cells from MSCs has been a challenge in the field of stem cell therapy of diabetes. Adipose tissue-derived mesenchymal stem cells (Ad-MSCs) have made their mark in regenerative medicine, and provide several advantages compared to other MSCs sources. Forkhead box protein O-1 (FOXO-1) is an important transcription factor for normal development of β-cells, yet its over expression in β-cells may cause glucose intolerance. In this study, we isolated, characterized Ad-MSCs from rat epididymal fat pads, differentiated these MSCs into insulin producing cells (IPCs) and studied the role of FOXO-1 in such differentiation. MATERIALS AND METHODS We examined the expression of FOXO-1 and its nuclear cytoplasmic localization in the generated IPCs. Afterwards we knocked down FOXO-1 using siRNA targeting FOXO-1 (siFOXO-1). The differentiated siFOXO-1 IPCs were compared to non-targeting siRNA (siNT) IPCs regarding expression of β-cell markers by qRT-PCR and western blotting, dithizone (DTZ) staining and glucose stimulated insulin secretion (GSIS). KEY FINDINGS Isolated Ad-MSCs exhibited all characteristics of MSCs and can generate IPCs. FOXO-1 was initially elevated during differentiation followed by a decline towards end of differentiation. FOXO-1 was dephosphorylated and localized to the nucleus upon differentiation into IPCs. Knock down of FOXO-1 improved the expression of β-cell markers in final differentiated IPCs, improved DTZ uptake and showed increased insulin secretion upon challenging with increased glucose concentration. SIGNIFICANCE These results portray FOXO-1 as a hindering factor of generation of IPCs whose down-regulation can generate more mature IPCs for MSCs therapy of diabetes mellitus.
Collapse
Affiliation(s)
- Mohamed M Kamal
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt; Health Research Center of Excellence, Drug Research and Development Group, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt.
| | - Reham A Ammar
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt; Health Research Center of Excellence, Drug Research and Development Group, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Dina H Kassem
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
2
|
Sepyani S, Momenzadeh S, Safabakhsh S, Nedaeinia R, Salehi R. Therapeutic approaches for Type 1 Diabetes: Promising cell-based approaches to achieve ultimate success. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:23-33. [PMID: 37977308 DOI: 10.1016/j.slasd.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/12/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Type 1 Diabetes mellitus (T1DM) is a chronic metabolic disorder characterized by pancreatic β-cells destruction. Despite substantial advances in T1DM treatment, lifelong exogenous insulin administration is the mainstay of treatments, and constant control of glucose levels is still a challenge. Endogenous insulin production by replacing insulin-producing cells is an alternative, but the lack of suitable donors is accounted as one of the main obstacles to its widespread application. The research and trials overview demonstrates that endogenous production of insulin has started to go beyond the deceased-derived to stem cells-derived insulin-producing cells. Several protocols have been developed over the past couple of years for generating insulin-producing cells (IPCs) from various stem cell types and reprogramming fully differentiated cells. A straightforward and quick method for achieving this goal is to investigate and apply the β-cell specific transcription factors as a direct strategy for IPCs generation. In this review, we emphasize the significance of transcription factors in IPCs development from different non-beta cell sources, and pertinent research underlies the marked progress in the methods for generating insulin-producing cells and application for Type 1 Diabetes treatment.
Collapse
Affiliation(s)
- Sahar Sepyani
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sedigheh Momenzadeh
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saied Safabakhsh
- Micronesian Institute for Disease Prevention and Research, 736 Route 4, Suite 103, Sinajana, GU 96910, United States
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rasoul Salehi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
3
|
Accili D, Du W, Kitamoto T, Kuo T, McKimpson W, Miyachi Y, Mukhanova M, Son J, Wang L, Watanabe H. Reflections on the state of diabetes research and prospects for treatment. Diabetol Int 2023; 14:21-31. [PMID: 36636157 PMCID: PMC9829952 DOI: 10.1007/s13340-022-00600-2] [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: 06/27/2022] [Accepted: 08/02/2022] [Indexed: 01/16/2023]
Abstract
Research on the etiology and treatment of diabetes has made substantial progress. As a result, several new classes of anti-diabetic drugs have been introduced in clinical practice. Nonetheless, the number of patients achieving glycemic control targets has not increased for the past 20 years. Two areas of unmet medical need are the restoration of insulin sensitivity and the reversal of pancreatic beta cell failure. In this review, we integrate research advances in transcriptional regulation of insulin action and pathophysiology of beta cell dedifferentiation with their potential impact on prospects of a durable "cure" for patients suffering from type 2 diabetes.
Collapse
Affiliation(s)
- Domenico Accili
- Department of Medicine and Berrie Diabetes Center, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY 10032 USA
| | - Wen Du
- Department of Medicine and Berrie Diabetes Center, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY 10032 USA
| | - Takumi Kitamoto
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba, Chiba 260-8670 Japan
| | - Taiyi Kuo
- Department of Neurobiology, Physiology, and Behavior, University of California at Davis, Davis, CA 95616 USA
| | - Wendy McKimpson
- Department of Medicine and Berrie Diabetes Center, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY 10032 USA
| | - Yasutaka Miyachi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Fukuoka Japan
| | - Maria Mukhanova
- Department of Medicine and Berrie Diabetes Center, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY 10032 USA
| | - Jinsook Son
- Department of Medicine and Berrie Diabetes Center, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY 10032 USA
| | - Liheng Wang
- Department of Medicine and Berrie Diabetes Center, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY 10032 USA
| | - Hitoshi Watanabe
- Department of Medicine and Berrie Diabetes Center, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY 10032 USA
| |
Collapse
|
4
|
Baafi K, March JC. Harnessing gut cells for functional insulin production: Strategies and challenges. BIOTECHNOLOGY NOTES (AMSTERDAM, NETHERLANDS) 2022; 4:7-13. [PMID: 39416909 PMCID: PMC11446352 DOI: 10.1016/j.biotno.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/30/2022] [Accepted: 11/30/2022] [Indexed: 10/19/2024]
Abstract
Reprogrammed glucose-responsive, insulin + cells ("β-like") exhibit the potential to bypass the hurdles of exogenous insulin delivery in treating diabetes mellitus. Current cell-based therapies-transcription factor regulation, biomolecule-mediated enteric signaling, and transgenics - have demonstrated the promise of reprogramming either mature or progenitor gut cells into surrogate "β-like" cells. However, there are predominant challenges impeding the use of gut "β-like" cells as clinical replacements for insulin therapy. Reprogrammed "β-like" gut cells, even those of enteroendocrine origin, mostly do not exhibit glucose - potentiated insulin secretion. Despite the exceptionally low conversion rate of gut cells into surrogate "β-like" cells, the therapeutic quantity of gut "β-like" cells needed for normoglycemia has not even been established. There is also a lingering uncertainty regarding the functionality and bioavailability of gut derived insulin. Herein, we review the strategies, challenges, and opportunities in the generation of functional, reprogrammed "β-like" cells.
Collapse
Affiliation(s)
- Kelvin Baafi
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | | |
Collapse
|
5
|
Towarnicki SG, Youngson NA, Corley SM, St. John JC, Melvin RG, Turner N, Morris MJ, Ballard JWO. Ancestral dietary change alters the development of Drosophila larvae through MAPK signalling. Fly (Austin) 2022; 16:299-311. [PMID: 35765944 PMCID: PMC9354765 DOI: 10.1080/19336934.2022.2088032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Studies in a broad range of animal species have revealed phenotypes that are caused by ancestral life experiences, including stress and diet. Ancestral dietary macronutrient composition and quantity (over- and under-nutrition) have been shown to alter descendent growth, metabolism and behaviour. Molecules have been identified in gametes that are changed by ancestral diet and are required for transgenerational effects. However, there is less understanding of the developmental pathways altered by inherited molecules during the period between fertilization and adulthood. To investigate this non-genetic inheritance, we exposed great grand-parental and grand-parental generations to defined protein to carbohydrate (P:C) dietary ratios. Descendent developmental timing was consistently faster in the period between the embryonic and pupal stages when ancestors had a higher P:C ratio diet. Transcriptional analysis revealed extensive and long-lasting changes to the MAPK signalling pathway, which controls growth rate through the regulation of ribosomal RNA transcription. Pharmacological inhibition of both MAPK and rRNA pathways recapitulated the ancestral diet-induced developmental changes. This work provides insight into non-genetic inheritance between fertilization and adulthood.
Collapse
Affiliation(s)
- Samuel G. Towarnicki
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Neil A. Youngson
- Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW, Australia,The Institute of Hepatology, The Foundation for Liver Research, London, UK
| | - Susan M. Corley
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Jus C. St. John
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Richard G. Melvin
- Department of Environment and Genetics, La Trobe University, Melbourne, VIC, Australia
| | - Nigel Turner
- The Institute of Hepatology, The Foundation for Liver Research, London, UK
| | - Margaret J. Morris
- The Institute of Hepatology, The Foundation for Liver Research, London, UK
| | - J. William O. Ballard
- Department of Environment and Genetics, La Trobe University, Melbourne, VIC, Australia,Department of Ecology, Environment and Evolution, School of Life Sciences, Victoria 3086, La Trobe University, Melbourne, VIC, Australia,CONTACT J. William O. Ballard Department of Environment and Genetics, SABE, La Trobe University, Bundoora, VIC3086, Australia
| |
Collapse
|
6
|
Kitamoto T, Lee YK, Sultana N, Watanabe H, McKimpson WM, Du W, Fan J, Diaz B, Lin HV, Leibel RL, Belvedere S, Accili D, Accili D. Chemical induction of gut β-like-cells by combined FoxO1/Notch inhibition as a glucose-lowering treatment for diabetes. Mol Metab 2022; 66:101624. [PMID: 36341906 PMCID: PMC9664469 DOI: 10.1016/j.molmet.2022.101624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE Lifelong insulin replacement remains the mainstay of type 1 diabetes treatment. Genetic FoxO1 ablation promotes enteroendocrine cell (EECs) conversion into glucose-responsive β-like cells. Here, we tested whether chemical FoxO1 inhibitors can generate β-like gut cells. METHODS We used Ngn3-or Villin-driven FoxO1 ablation to capture the distinctive developmental effects of FoxO1 on EEC pool. We combined FoxO1 ablation with Notch inhibition to enhance the expansion of EEC pool. We tested the ability of an orally available small molecule of FoxO1 inhibitor, Cpd10, to phenocopy genetic ablation of FoxO1. We evaluated the therapeutic impact of genetic ablation or chemical inhibition of FoxO1 on insulin-deficient diabetes in Ins2Akita/+ mice. RESULTS Pan-intestinal epithelial FoxO1 ablation expanded the EEC pool, induced β-like cells, and improved glucose tolerance in Ins2Akita/+ mice. This genetic effect was phenocopied by Cpd10. Cpd10 induced β-like cells that released insulin in response to glucose in gut organoids, and this effect was enhanced by the Notch inhibitor, DBZ. In Ins2Akita/+ mice, a five-day course of either Cpd10 or DBZ induced intestinal insulin-immunoreactive β-like cells, lowered glycemia, and increased plasma insulin levels without apparent adverse effects. CONCLUSION These results provide proof of principle of gut cell conversion into β-like cells by a small molecule FoxO1 inhibitor, paving the way for clinical applications.
Collapse
Affiliation(s)
- Takumi Kitamoto
- Department of Medicine and Columbia University, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA; Chiba University Graduate School of Medicine, Chiba, Japan, 2608670.
| | | | - Nishat Sultana
- Department of Pediatrics Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Hitoshi Watanabe
- Department of Medicine and Columbia University, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA
| | - Wendy M McKimpson
- Department of Medicine and Columbia University, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA
| | - Wen Du
- Department of Medicine and Columbia University, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA
| | - Jason Fan
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami, Miami, FL, 33146, USA
| | - Bryan Diaz
- Department of Pediatrics Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Hua V Lin
- BioFront Therapeutics, Beijing, China
| | - Rudolph L Leibel
- Department of Pediatrics Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | - Domenico Accili
- Department of Medicine and Columbia University, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA
| | | |
Collapse
|
7
|
Nie J, Liao W, Zhang Z, Zhang M, Wen Y, Capanoglu E, Sarker MMR, Zhu R, Zhao C. A 3D co-culture intestinal organoid system for exploring glucose metabolism. Curr Res Food Sci 2022; 6:100402. [DOI: 10.1016/j.crfs.2022.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/02/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
|
8
|
An Association between Insulin Resistance and Neurodegeneration in Zebrafish Larval Model ( Danio rerio). Int J Mol Sci 2022; 23:ijms23158290. [PMID: 35955446 PMCID: PMC9368350 DOI: 10.3390/ijms23158290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Type 2 diabetes mellitus has recently been identified as a mediator of neurodegeneration. However, the molecular mechanisms have not been clearly elucidated. We aimed to investigate insulin resistance associated with neurodegenerative events in zebrafish larvae. Methods: Larvae aged 72 h-post-fertilization (hpf) were induced to insulin resistance by immersion in 250 nM insulin and were then reinduced with 100 nM insulin at 96 hpf. This model was validated by a glucose levels assay, qPCR analysis of selected genes (akt, pepck, zglut3 and claudin-5a) and Oil Red-O (ORO) staining of the yolk sac for lipid distribution. The association of insulin resistance and neurodegeneration was validated by malondialdehyde (MDA), glutathione (GSH) assays, and by integrating next-generation sequencing with database for annotation, visualization and integrated discovery (DAVID). Results: There was a significant increase in glucose levels at 180 min in the insulin-resistant group. However, it decreased at 400 min after the re-challenge. Insulin-signaling mediators, akt and pepck, were showed significantly downregulated up to 400 min after insulin immersion (p < 0.05). Meanwhile, claudin-5a assessed blood−brain barrier (BBB) integrity and showed significant deterioration after 400 min of post-insulin immersion. ORO staining remarked the increase in yolk sac size in the insulin-resistant group. After the confirmation of insulin resistance, MDA levels increased significantly in the insulin-resistant group compared to the control group in the following parameters. Furthermore, dysregulated MAPK- and Wnt/Ca2+-signaling pathways were observed in the insulin-resistant group, disrupting energy metabolism and causing BBB injury. Conclusions: We conclude that the insulin-resistant zebrafish larvae alter the metabolic physiology associated with neurodegeneration.
Collapse
|
9
|
Yue J, Aobulikasimu A, Sun W, Liu S, Xie W, Sun W. Targeted regulation of FoxO1 in chondrocytes prevents age-related osteoarthritis via autophagy mechanism. J Cell Mol Med 2022; 26:3075-3082. [PMID: 35560791 PMCID: PMC9170816 DOI: 10.1111/jcmm.17319] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/17/2022] [Accepted: 03/30/2022] [Indexed: 11/30/2022] Open
Abstract
Autophagy is designated as a biological recycling process to maintain cellular homeostasis by the sequestration of damaged proteins and organelles in plasma and cargo delivery to lysosomes for degradation and reclamation. This organelle recycling process promotes chondrocyte homeostasis and has been previously implicated in osteoarthritis (OA). Autophagy is widely involved in regulating chondrocyte degeneration markers such as MMPs, ADAMSTs and Col10 in chondrocytes. The critical autophagy‐related (ATG) proteins have now been considered the protective factor against late‐onset OA. The current research field proposes that the autophagic pathway is closely related to chondrocyte activity. However, the mechanism is complex yet needs precise elaboration. This review concluded that FoxO1, a forkhead O family protein, which is a decisive mediator of autophagy, facilitates the pathological process of osteoarthritis. Diverse mechanisms regulate the activity of FoxO1 and promote the initiation of autophagy, including the prominent AMPK and Sirt‐2 cellular pathways. FoxO1 transactive is regulated by phosphorylation and acetylation processes, which modulates the downstream ATGs expression. Furthermore, FoxO1 induces autophagy by directly interacting with ATGs proteins, which control the formation of autophagosomes and lysosomes fusion. This review will discuss cutting‐edge evidence that the FoxO–autophagy pathway plays an essential regulator in the pathogenesis of osteoarthritis.
Collapse
Affiliation(s)
- Jiaji Yue
- Department of Bone and Joint Surgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Aikebaier Aobulikasimu
- Department of Bone and Joint Surgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Weichao Sun
- Department of Bone and Joint Surgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Shuyu Liu
- Department of Bone and Joint Surgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Wei Xie
- Department of Bone and Joint Surgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Wei Sun
- Department of Bone and Joint Surgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
10
|
Li X, Wan T, Li Y. Role of FoxO1 in regulating autophagy in type 2 diabetes mellitus (Review). Exp Ther Med 2021; 22:707. [PMID: 34007316 PMCID: PMC8120662 DOI: 10.3892/etm.2021.10139] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a major chronic disease that is characterized by pancreatic β-cell dysfunction and insulin resistance. Autophagy is a highly conserved intracellular recycling pathway and is involved in regulating intracellular homeostasis. Transcription factor Forkhead box O1 (FoxO1) also regulates fundamental cellular processes, including cell differentiation, metabolism and apoptosis, and proliferation to cellular stress. Increasing evidence suggest that autophagy and FoxO1 are involved in the pathogenesis of T2DM, including β-cell viability, apoptosis, insulin secretion and peripheral insulin resistance. Recent studies have demonstrated that FoxO1 improves insulin resistance by regulating target tissue autophagy. The present review summarizes current literature on the role of autophagy and FoxO1 in T2DM. The participation of FoxO1 in the development and occurrence of T2DM via autophagy is also discussed.
Collapse
Affiliation(s)
- Xiudan Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Tingting Wan
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yanbo Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
11
|
Direct suppression of human islet dedifferentiation, progenitor genes, but not epithelial to mesenchymal transition by liraglutide. Heliyon 2020; 6:e04951. [PMID: 32995630 PMCID: PMC7501427 DOI: 10.1016/j.heliyon.2020.e04951] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/03/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022] Open
Abstract
β-cell dedifferentiation has been accounted as one of the major mechanisms for β-cell failure; thus, is a cause to diabetes. We study direct impacts of liraglutide treatment on ex vivo human dedifferentiated islets, and its effects on genes important in endocrine function, progenitor states, and epithelial mesenchymal transition (EMT). Human islets from non-diabetic donors, were purified and incubated until day 1 and day 4, and were determined insulin contents, numbers of insulin (INS+) and glucagon (GCG+) cells. The islets from day 3 to day 7 were treated with diabetic drugs, the long acting GLP-1 receptor agonist, liraglutide. As observed in pancreatic islets of type 2 diabetic patients, ex vivo dedifferentiated islets showed more than 50% reduced insulin contents while number of glucagon increased from 10% to about 20%. β-cell specific genes: PDX1, MAFA, as well as β-cell functional markers: GLUT1 and SUR1, were significantly depleted more than 40%. Notably, we found increased levels of glucagon regulator, ARX and pre-glucagon transcripts, and remarkably upregulated progenitor expressions: NEUROG3 and ALDH1A identified as β-cell dysfunction markers in diabetic models. Hyperglucagonemia was often observed in type 2 patients that could lead to over production of gluconeogenesis by the liver. Liraglutide treatments resulted in decreased number of GCG+ cells, increased numbers of GLP-1 positive cells but did not alter elevated levels of EMT marker genes: ACTA2, CDH-2, SNAIL2, and VIM. These effects of liraglutide were blunted when FOXO1 transcripts were depleted. This work illustrates that ex vivo human isolated islets can be used as a tool to study different aspects of β-cell dedifferentiation. Our novel finding suggests a role of GLP-1 pathway in beta-cell maintenance in FOXO1-dependent manner. Importantly, dedifferentiated islets ex vivo is a useful model that can be utilized to verify the actions of potential drugs to diabetic β-cell failure.
Collapse
|
12
|
Liu Z, Sun F, Liu Z, Wang X, Jin M, Mao J, Wu Q, Yan S, Xu K, Wang K, Hu S. Effect of Sleeve Gastrectomy on Glycometabolism via Forkhead Box O1 (FoxO1)/Lipocalin-2 (LCN2) Pathway. Med Sci Monit 2020; 26:e927458. [PMID: 32845875 PMCID: PMC7780888 DOI: 10.12659/msm.927458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The mechanism by which sleeve gastrectomy (SG) improves glycometabolism has remained unclear so far. Increasing evidence has demonstrated that bone is a regulator of glucose metabolism, and osteoblast-derived forkhead box O1 (FoxO1) and lipocalin-2 (LCN2) are regulators of energy metabolism. The aim of this study was to investigate whether the FOXO1/LCN2 signaling pathway is involved in the anti-diabetic effect of SG. MATERIAL AND METHODS Insulin resistance was induced in Wistar rats, which were then intraperitoneally injected with streptozotocin to induce a type 2 diabetic state. Levels of fasting blood glucose, serum insulin, HbA1c, and LCN2 were analyzed at corresponding time points after SG and sham surgeries. The expressions of FOXO1, LCN2, and the melanocortin 4 receptor (MC4R) in bone and hypothalamus were detected by immunofluorescence. FOXO1 siRNA was applied to downregulate FOXO1 expression in osteoblasts of rats. The influence of FOXO1 gene on expression of LCN2 was investigated in cultured osteoblasts by western blot and PCR. RESULTS Glucose metabolism in the SG group was significantly improved. The LCN2 expression in bone in the SG group was higher than that in the sham group, whereas FOXO1 expression in the SG group was lower than that in the sham group. The binding rate of LCN2 and MC4R in the hypothalamus was also higher in the SG group compared with that in the sham group. The downregulation of FOXO1 expression in osteoblasts was accompanied by upregulation of LCN2 expression. CONCLUSIONS These results suggest that the FOXO1/LCN2 signaling pathway participates in the anti-diabetic effect of SG.
Collapse
Affiliation(s)
- Zhi Liu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Fuyun Sun
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Zitian Liu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Xiaoyang Wang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Mingxin Jin
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Jiajia Mao
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Qunzheng Wu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Shaohua Yan
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Kai Xu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Kexin Wang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Sanyuan Hu
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
13
|
Sasaki B, Uemoto S, Kawaguchi Y. Transient FOXO1 inhibition in pancreatic endoderm promotes the generation of NGN3+ endocrine precursors from human iPSCs. Stem Cell Res 2020; 44:101754. [PMID: 32179491 DOI: 10.1016/j.scr.2020.101754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 01/24/2020] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
In the multi-step differentiation protocol used to generate pancreatic endocrine cells from human pluripotent stem cells, the induction of NGN3+ endocrine precursors from the PDX1+/NKX6.1+ pancreatic endoderm is crucial for efficient endocrine cell production. Here, we demonstrate that transient, not prolonged FOXO1 inhibition results in enhanced NGN3+ endocrine precursors and hormone-producing cell production. FOXO1 inhibition does not directly induce NGN3 expression but stimulates PDX1+/NKX6.1+ cell proliferation. NOTCH activity, whose suppression is important for Ngn3 expression, is not suppressed but Wnt signaling is stimulated by FOXO1 inhibition. Reversely, Wnt inhibition suppresses the effects of FOXO1 inhibitor. These findings indicate that FOXO1 and Wnt are involved in regulating the proliferation of PDX1+/NKX6.1+ pancreatic endoderm that gives rise to NGN3+ endocrine precursors.
Collapse
Affiliation(s)
- Ben Sasaki
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Department of Hepato-Biliary-Pancreatic Surgery and Transplantation, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shinji Uemoto
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yoshiya Kawaguchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
14
|
Lee KI, Choi S, Matsuzaki T, Alvarez-Garcia O, Olmer M, Grogan SP, D'Lima DD, Lotz MK. FOXO1 and FOXO3 transcription factors have unique functions in meniscus development and homeostasis during aging and osteoarthritis. Proc Natl Acad Sci U S A 2020; 117:3135-3143. [PMID: 31980519 PMCID: PMC7022148 DOI: 10.1073/pnas.1918673117] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The objective of this study was to examine FoxO expression and FoxO function in meniscus. In menisci from human knee joints with osteoarthritis (OA), FoxO1 and 3 expression were significantly reduced compared with normal menisci from young and old normal donors. The expression of FoxO1 and 3 was also significantly reduced in mouse menisci during aging and OA induced by surgical meniscus destabilization or mechanical overuse. Deletion of FoxO1 and combined FoxO1, 3, and 4 deletions induced abnormal postnatal meniscus development in mice and these mutant mice spontaneously displayed meniscus pathology at 6 mo. Mice with Col2Cre-mediated deletion of FoxO3 or FoxO4 had normal meniscus development but had more severe aging-related damage. In mature AcanCreERT2 mice, the deletion of FoxO1, 3, and 4 aggravated meniscus lesions in all experimental OA models. FoxO deletion suppressed autophagy and antioxidant defense genes and altered several meniscus-specific genes. Expression of these genes was modulated by adenoviral FoxO1 in cultured human meniscus cells. These results suggest that FoxO1 plays a key role in meniscus development and maturation, and both FoxO1 and 3 support homeostasis and protect against meniscus damage in response to mechanical overuse and during aging and OA.
Collapse
Affiliation(s)
- Kwang Il Lee
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Sungwook Choi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Department of Orthopaedic Surgery, Jeju National University College of Medicine, 63243 Jeju, South Korea
| | - Tokio Matsuzaki
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Oscar Alvarez-Garcia
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Merissa Olmer
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Shawn P Grogan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Darryl D D'Lima
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Martin K Lotz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037;
| |
Collapse
|
15
|
FOXO1 and FOXO3 transcription factors have unique functions in meniscus development and homeostasis during aging and osteoarthritis. Proc Natl Acad Sci U S A 2020. [PMID: 31980519 DOI: 10.1073/pnas.1918673117.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The objective of this study was to examine FoxO expression and FoxO function in meniscus. In menisci from human knee joints with osteoarthritis (OA), FoxO1 and 3 expression were significantly reduced compared with normal menisci from young and old normal donors. The expression of FoxO1 and 3 was also significantly reduced in mouse menisci during aging and OA induced by surgical meniscus destabilization or mechanical overuse. Deletion of FoxO1 and combined FoxO1, 3, and 4 deletions induced abnormal postnatal meniscus development in mice and these mutant mice spontaneously displayed meniscus pathology at 6 mo. Mice with Col2Cre-mediated deletion of FoxO3 or FoxO4 had normal meniscus development but had more severe aging-related damage. In mature AcanCreERT2 mice, the deletion of FoxO1, 3, and 4 aggravated meniscus lesions in all experimental OA models. FoxO deletion suppressed autophagy and antioxidant defense genes and altered several meniscus-specific genes. Expression of these genes was modulated by adenoviral FoxO1 in cultured human meniscus cells. These results suggest that FoxO1 plays a key role in meniscus development and maturation, and both FoxO1 and 3 support homeostasis and protect against meniscus damage in response to mechanical overuse and during aging and OA.
Collapse
|
16
|
FCoR-Foxo1 Axis Regulates α-Cell Mass through Repression of Arx Expression. iScience 2019; 23:100798. [PMID: 31923647 PMCID: PMC6951314 DOI: 10.1016/j.isci.2019.100798] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 11/10/2019] [Accepted: 12/19/2019] [Indexed: 01/27/2023] Open
Abstract
Pancreatic endocrine cell development into differentiated α- and β-cells is highly regulated and involves multiple transcription factors. However, the mechanisms behind the determination of α- and β-cell masses remains unclear. We previously identified Foxo1 CoRepressor (FCoR), which inhibits Foxo1 by acetylation. Here we demonstrate that Fcor-knockout mice (FcorKO) exhibit significantly increased α-cell mass, expression of the master α-cell regulatory transcription factor Aristaless-related homeobox (Arx), which can be normalized by β-cell-specific FCoR overexpression (FcorKO-βFcor), and exhibit β-to-α-cell conversion. Compared with FcorKO, β-cell-specific Foxo1 knockout in the FcorKO (DKO) led to decreased Arx expression and α-cell mass. Foxo1 binding to Arx promoter led to DNA methyltransferase 3a (Dnmt3a) dissociation, Arx promoter hypomethylation, and increased Arx expression. In contrast, FCoR suppressed Arx through Foxo1 inhibition and Dnmt3a recruitment to Arx promoter and increased Arx promoter methylation. Our findings suggest that the FCoR-Foxo1 axis regulates pancreatic α-cell mass by suppressing Arx expression.
Collapse
|
17
|
Dayem AA, Lee SB, Kim K, Lim KM, Jeon TI, Cho SG. Recent advances in organoid culture for insulin production and diabetes therapy: methods and challenges. BMB Rep 2019. [PMID: 30940326 PMCID: PMC6549913 DOI: 10.5483/bmbrep.2019.52.5.089] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Breakthroughs in stem cell technology have contributed to disease modeling and drug screening via organoid technology. Organoid are defined as three-dimensional cellular aggregations derived from adult tissues or stem cells. They recapitulate the intricate pattern and functionality of the original tissue. Insulin is secreted mainly by the pancreatic β cells. Large-scale production of insulin-secreting β cells is crucial for diabetes therapy. Here, we provide a brief overview of organoids and focus on recent advances in protocols for the generation of pancreatic islet organoids from pancreatic tissue or pluripotent stem cells for insulin secretion. The feasibility and limitations of organoid cultures derived from stem cells for insulin production will be described. As the pancreas and gut share the same embryological origin and produce insulin, we will also discuss the possible application of gut organoids for diabetes therapy. Better understanding of the challenges associated with the current protocols for organoid culture facilitates development of scalable organoid cultures for applications in biomedicine.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Soo Bin Lee
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Kyeongseok Kim
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Kyung Min Lim
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Tak-Il Jeon
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Ssang-Goo Cho
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
18
|
Identification of C2CD4A as a human diabetes susceptibility gene with a role in β cell insulin secretion. Proc Natl Acad Sci U S A 2019; 116:20033-20042. [PMID: 31527256 DOI: 10.1073/pnas.1904311116] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Fine mapping and validation of genes causing β cell failure from susceptibility loci identified in type 2 diabetes genome-wide association studies (GWAS) poses a significant challenge. The VPS13C-C2CD4A-C2CD4B locus on chromosome 15 confers diabetes susceptibility in every ethnic group studied to date. However, the causative gene is unknown. FoxO1 is involved in the pathogenesis of β cell dysfunction, but its link to human diabetes GWAS has not been explored. Here we generated a genome-wide map of FoxO1 superenhancers in chemically identified β cells using 2-photon live-cell imaging to monitor FoxO1 localization. When parsed against human superenhancers and GWAS-derived diabetes susceptibility alleles, this map revealed a conserved superenhancer in C2CD4A, a gene encoding a β cell/stomach-enriched nuclear protein of unknown function. Genetic ablation of C2cd4a in β cells of mice phenocopied the metabolic abnormalities of human carriers of C2CD4A-linked polymorphisms, resulting in impaired insulin secretion during glucose tolerance tests as well as hyperglycemic clamps. C2CD4A regulates glycolytic genes, and notably represses key β cell "disallowed" genes, such as lactate dehydrogenase A We propose that C2CD4A is a transcriptional coregulator of the glycolytic pathway whose dysfunction accounts for the diabetes susceptibility associated with the chromosome 15 GWAS locus.
Collapse
|
19
|
Qi H, Yao L, Liu Q. MicroRNA-96 regulates pancreatic β cell function under the pathological condition of diabetes mellitus through targeting Foxo1 and Sox6. Biochem Biophys Res Commun 2019; 519:294-301. [PMID: 31506178 DOI: 10.1016/j.bbrc.2019.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 09/01/2019] [Indexed: 01/13/2023]
Abstract
To elucidate the potential function of microRNA-96 in protecting pancreatic β cell function under the pathological condition of T2DM and the underlying mechanism. Relative levels of microRNA-96 and genes associated with β cell function in the in vivo and in vitro T2DM and obesity models were detected by qRT-PCR. Insulin functions, including fasting blood glucose, plasma insulin, HOMA-IR, HOMA-%b, glucose tolerance and insulin tolerance, were assessed in microRNA-96 KO mice and wild-type mice fed with normal diet or high-fat diet. Downstream targets of microRNA-96 were verified by dual-luciferase reporter gene assay. Finally, regulatory effects of microRNA-96 on proliferation and apoptosis of MIN6 cells were determined. MicroRNA-96 was upregulated in mice fed with high-fat diet, db/db mice, high-level glucose-treated cells, TNF-α-treated cells, pancreatic cells isolated from the obesity and T2DM patients. Increased fasting blood glucose and HOMA-IR, as well as decreased plasma insulin and HOMA-%b were observed in microRNA-96 KO mice. IPGTT and IPITT results indicated that knockout of microRNA-96 led to pancreatic β cell dysfunction under the pathological condition of T2DM. Dual-luciferase reporter gene assay confirmed that microRNA-96 could bind Foxo1 and Sox6. MicroRNA-96 negatively regulated Foxo1 and Sox6 levels. Moreover, overexpression of microRNA-96 promoted proliferative ability and inhibited apoptosis in MIN6 cells. Relative levels of Pdx1, Nkx6.1, Cyclin D1 and Cyclin E1 were upregulated in MIN6 cells overexpressing microRNA-96. Opposite results were obtained after knockdown of microRNA-96 in MIN6 cells. MicroRNA-96 is upregulated in pancreatic β cells under the pathological condition of T2DM. Overexpression of microRNA-96 promotes proliferative ability and inhibits apoptosis in pancreatic β cells through targeting Foxo1 and Sox6.
Collapse
Affiliation(s)
- Huimeng Qi
- Department of General Practice, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, 110001, China.
| | - Qiang Liu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
20
|
Zhang K, Guo X, Yan H, Wu Y, Pan Q, Shen JZ, Li X, Chen Y, Li L, Qi Y, Xu Z, Xie W, Zhang W, Threadgill D, He L, Villarreal D, Sun Y, White MF, Zheng H, Guo S. Phosphorylation of Forkhead Protein FoxO1 at S253 Regulates Glucose Homeostasis in Mice. Endocrinology 2019; 160:1333-1347. [PMID: 30951171 PMCID: PMC6482038 DOI: 10.1210/en.2018-00853] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 03/29/2019] [Indexed: 01/04/2023]
Abstract
The transcription factor forkhead box O1 (FoxO1) is a key mediator in the insulin signaling pathway and controls multiple physiological functions, including hepatic glucose production (HGP) and pancreatic β-cell function. We previously demonstrated that S256 in human FOXO1 (FOXO1-S256), equivalent to S253 in mouse FoxO1 (FoxO1-S253), is a key phosphorylation site mediating the effect of insulin as a target of protein kinase B on suppression of FOXO1 activity and expression of target genes responsible for gluconeogenesis. Here, we investigated the role of FoxO1-S253 phosphorylation in control of glucose homeostasis in vivo by generating global FoxO1-S253A/A knockin mice, in which FoxO1-S253 alleles were replaced with alanine (A substitution) blocking FoxO1-S253 phosphorylation. FoxO1-S253A/A mice displayed mild increases in feeding blood glucose and insulin levels but decreases in fasting blood glucose and glucagon concentrations, as well as a reduction in the ratio of pancreatic α-cells/β-cells per islet. FoxO1-S253A/A mice exhibited a slight increase in energy expenditure but barely altered food intake and glucose uptake among tissues. Further analyses revealed that FoxO1-S253A/A enhances FoxO1 nuclear localization and promotes the effect of glucagon on HGP. We conclude that dephosphorylation of S253 in FoxO1 may reflect a molecular basis of pancreatic plasticity during the development of insulin resistance.
Collapse
Affiliation(s)
- Kebin Zhang
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Xiaoqin Guo
- Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Hui Yan
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Yuxin Wu
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
- Queens University Belfast School of Biological Sciences, Belfast, United Kingdom
| | - Quan Pan
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - James Zheng Shen
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Xiaopeng Li
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Yunmei Chen
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Ling Li
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Yajuan Qi
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Zihui Xu
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Wei Xie
- Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Weiping Zhang
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - David Threadgill
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Ling He
- Division of Endocrinology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Daniel Villarreal
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Yuxiang Sun
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Morris F White
- Division of Endocrinology, Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Hongting Zheng
- Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shaodong Guo
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
- Correspondence: Shaodong Guo, PhD, Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, 123A Cater-Mattil Hall, College Station, Texas 77843. E-mail:
| |
Collapse
|
21
|
Dayem AA, Lee SB, Kim K, Lim KM, Jeon TI, Cho SG. Recent advances in organoid culture for insulin production and diabetes therapy: methods and challenges. BMB Rep 2019; 52:295-303. [PMID: 30940326 PMCID: PMC6549913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Indexed: 10/13/2023] Open
Abstract
Breakthroughs in stem cell technology have contributed to disease modeling and drug screening via organoid technology. Organoid are defined as three-dimensional cellular aggregations derived from adult tissues or stem cells. They recapitulate the intricate pattern and functionality of the original tissue. Insulin is secreted mainly by the pancreatic β cells. Large-scale production of insulin-secreting β cells is crucial for diabetes therapy. Here, we provide a brief overview of organoids and focus on recent advances in protocols for the generation of pancreatic islet organoids from pancreatic tissue or pluripotent stem cells for insulin secretion. The feasibility and limitations of organoid cultures derived from stem cells for insulin production will be described. As the pancreas and gut share the same embryological origin and produce insulin, we will also discuss the possible application of gut organoids for diabetes therapy. Better understanding of the challenges associated with the current protocols for organoid culture facilitates development of scalable organoid cultures for applications in biomedicine. [BMB Reports 2019; 52(5): 295-303].
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029,
Korea
| | - Soo Bin Lee
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029,
Korea
| | - Kyeongseok Kim
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029,
Korea
| | - Kyung Min Lim
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029,
Korea
| | - Tak-il Jeon
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029,
Korea
| | - Ssang-Goo Cho
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029,
Korea
| |
Collapse
|
22
|
Abstract
OBJECTIVES Menin, a chromatin binding protein, interacts with various epigenetic regulators to regulate gene transcription, whereas forkhead box protein O1 (FOXO1) is a transcription factor that can be regulated by multiple signaling pathways. Both menin and FOXO1 are crucial regulators of β-cell function and metabolism; however, whether or how they interplay to regulate β cells is not clear. METHODS To examine whether menin affects expression of FOXO1, we ectopically expressed menin complementary DNA and small hairpin RNA targeting menin via a retroviral vector in INS-1 cells. Western blotting was used to analyze protein levels. RESULTS Our current work shows that menin increases the expression of FOXO1. Menin stabilizes FOXO1 protein level in INS-1 cells, as shown by increased half-life of FOXO1 by menin expression. Moreover, menin represses ubiquitination of FOXO1 protein and AKT phosphorylation, We found that menin stabilizes FOXO1 by repressing FOXO1 degradation mediated by S-phase kinase-associated protein 2 (Skp2), an E3 ubiquitin ligase, promoting caspase 3 activation and apoptosis. CONCLUSIONS Because FOXO1 upregulates the menin gene transcription, our findings unravel a crucial menin and FOXO1 interplay, with menin and FOXO1 upregulating their expression reciprocally, forming a positive feedback loop to sustain menin and FOXO1 expression.
Collapse
|
23
|
Accili D. Insulin Action Research and the Future of Diabetes Treatment: The 2017 Banting Medal for Scientific Achievement Lecture. Diabetes 2018; 67:1701-1709. [PMID: 30135131 PMCID: PMC6110318 DOI: 10.2337/dbi18-0025] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diabetes is caused by combined abnormalities in insulin production and action. The pathophysiology of these defects has been studied extensively and is reasonably well understood. Their causes are elusive and their manifestations pleiotropic, likely reflecting the triple threat of genes, environment, and lifestyle. Treatment, once restricted to monotherapy with secretagogues or insulin, now involves complex combinations of expensive regimens that stem the progression but do not fundamentally alter the underlying causes of the disease. As advances in our understanding of insulin action and β-cell failure reach a critical stage, here I draw on lessons learned from our research on insulin regulation of gene expression and pancreatic β-cell dedifferentiation to address the question of how we can translate this exciting biology into mechanism-based interventions to reverse the course of diabetes.
Collapse
MESH Headings
- Animals
- Awards and Prizes
- Cell Dedifferentiation/drug effects
- Cell Transdifferentiation/drug effects
- Cellular Reprogramming/drug effects
- Combined Modality Therapy/adverse effects
- Diabetes Complications/prevention & control
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/therapy
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/therapy
- Drug Design
- Drug Therapy, Combination/adverse effects
- Enteroendocrine Cells/drug effects
- Enteroendocrine Cells/metabolism
- Enteroendocrine Cells/pathology
- Forkhead Transcription Factors/antagonists & inhibitors
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Humans
- Hypoglycemic Agents/adverse effects
- Hypoglycemic Agents/chemistry
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Insulin/adverse effects
- Insulin/metabolism
- Insulin/pharmacology
- Insulin/therapeutic use
- Insulin Resistance
- Insulin Secretion
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Models, Biological
Collapse
Affiliation(s)
- Domenico Accili
- Naomi Berrie Diabetes Center and Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| |
Collapse
|
24
|
Tsuda M, Fukuda A, Roy N, Hiramatsu Y, Leonhardt L, Kakiuchi N, Hoyer K, Ogawa S, Goto N, Ikuta K, Kimura Y, Matsumoto Y, Takada Y, Yoshioka T, Maruno T, Yamaga Y, Kim GE, Akiyama H, Ogawa S, Wright CV, Saur D, Takaori K, Uemoto S, Hebrok M, Chiba T, Seno H. The BRG1/SOX9 axis is critical for acinar cell-derived pancreatic tumorigenesis. J Clin Invest 2018; 128:3475-3489. [PMID: 30010625 PMCID: PMC6063489 DOI: 10.1172/jci94287] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 05/23/2018] [Indexed: 02/06/2023] Open
Abstract
Chromatin remodeler Brahma related gene 1 (BRG1) is silenced in approximately 10% of human pancreatic ductal adenocarcinomas (PDAs). We previously showed that BRG1 inhibits the formation of intraductal pancreatic mucinous neoplasm (IPMN) and that IPMN-derived PDA originated from ductal cells. However, the role of BRG1 in pancreatic intraepithelial neoplasia-derived (PanIN-derived) PDA that originated from acinar cells remains elusive. Here, we found that exclusive elimination of Brg1 in acinar cells of Ptf1a-CreER; KrasG12D; Brg1fl/fl mice impaired the formation of acinar-to-ductal metaplasia (ADM) and PanIN independently of p53 mutation, while PDA formation was inhibited in the presence of p53 mutation. BRG1 bound to regions of the Sox9 promoter to regulate its expression and was critical for recruitment of upstream regulators, including PDX1, to the Sox9 promoter and enhancer in acinar cells. SOX9 expression was downregulated in BRG1-depleted ADMs/PanINs. Notably, Sox9 overexpression canceled this PanIN-attenuated phenotype in KBC mice. Furthermore, Brg1 deletion in established PanIN by using a dual recombinase system resulted in regression of the lesions in mice. Finally, BRG1 expression correlated with SOX9 expression in human PDAs. In summary, BRG1 is critical for PanIN initiation and progression through positive regulation of SOX9. Thus, the BRG1/SOX9 axis is a potential target for PanIN-derived PDA.
Collapse
Affiliation(s)
- Motoyuki Tsuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Nilotpal Roy
- Diabetes Center, Department of Medicine, UCSF, San Francisco, California, USA
| | - Yukiko Hiramatsu
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Laura Leonhardt
- Diabetes Center, Department of Medicine, UCSF, San Francisco, California, USA
| | - Nobuyuki Kakiuchi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Pathology and Tumor Biology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kaja Hoyer
- Department of Pathology and Tumor Biology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Hematology, Oncology and Tumorimmunology, Charite–Universitätsmedizin Berlin, Berlin, Germany
| | - Satoshi Ogawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Norihiro Goto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kozo Ikuta
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshito Kimura
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshihide Matsumoto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yutaka Takada
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takuto Yoshioka
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takahisa Maruno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuichi Yamaga
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Grace E. Kim
- Department of Pathology, UCSF, San Francisco, California, USA
| | | | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Christopher V. Wright
- Program in Developmental Biology and Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Dieter Saur
- Department of Internal Medicine II, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Kyoichi Takaori
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shinji Uemoto
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, UCSF, San Francisco, California, USA
| | - Tsutomu Chiba
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Kansai Electric Power Hospital, Osaka, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
25
|
Jiang Z, Shi D, Tu Y, Tian J, Zhang W, Xing B, Wang J, Liu S, Lou J, Gustafsson JÅ, Hua X, Ma X. Human Proislet Peptide Promotes Pancreatic Progenitor Cells to Ameliorate Diabetes Through FOXO1/Menin-Mediated Epigenetic Regulation. Diabetes 2018; 67:1345-1355. [PMID: 29716892 DOI: 10.2337/db17-0885] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 04/17/2018] [Indexed: 11/13/2022]
Abstract
We investigated how human proislet peptide (HIP) regulates differentiation of human fetus-derived pancreatic progenitor cells (HFPPCs) and explored the potential link between HIP signaling and the menin pathway, which is key to regulating pancreatic islet differentiation. The data show that HIP promoted expression of proislet transcription factors (TFs), including PDX-1, MAFA, and NKX6.1, as well as other maturation markers of β-cells, such as insulin, GLUT2, KIR6.2, SUR1, and VDCC. Moreover, HIP increased insulin content and promoted the ability of HFPPCs to normalize blood glucose in diabetic mice. HIP inhibited the TF FOXO1 by increasing AKT-mediated phosphorylation. HIP-induced repression of FOXO1 suppressed menin expression, leading to reducing menin binding to the promoter of the three key proislet TFs, decreasing recruitment of H3K9 methyltransferase SUV39H1, and thus reducing repressive H3K9me3 at the promoter. These coordinated actions lead to increased expression of the proislet TFs, resulting in induction of HFPPC differentiation. Consistently, constitutive activation of FOXO1 blocks HIP-induced transcription of these TFs. Together, these studies unravel the crucial role of the HIP/AKT/FOXO/menin axis in epigenetically controlling expression of proislet TFs, regulating the differentiation of HFPPCs, and normalizing blood glucose in diabetic mice.
Collapse
Affiliation(s)
- Zongzhe Jiang
- Shenzhen University School of Medicine, Shenzhen, China
| | - Diwen Shi
- Shenzhen University School of Medicine, Shenzhen, China
| | - Yifan Tu
- Shenzhen University School of Medicine, Shenzhen, China
| | - Jingjing Tian
- Shenzhen University School of Medicine, Shenzhen, China
| | - Wenjian Zhang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Bowen Xing
- Shenzhen University School of Medicine, Shenzhen, China
| | - Jihua Wang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Suhuan Liu
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jinning Lou
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry and Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX
| | - Xianxin Hua
- Shenzhen University School of Medicine, Shenzhen, China
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Xiaosong Ma
- Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
26
|
Cao W, Li M, Wu T, Feng F, Feng T, Xu Y, Sun C. αMSH prevents ROS-induced apoptosis by inhibiting Foxo1/mTORC2 in mice adipose tissue. Oncotarget 2018; 8:40872-40884. [PMID: 28388573 PMCID: PMC5522219 DOI: 10.18632/oncotarget.16606] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/02/2017] [Indexed: 12/16/2022] Open
Abstract
Alpha-melanocyte stimulating hormone (αMSH) is an important adenohypophysis polypeptide hormone that regulates body metabolic status. To date, it is well known that the disorder of hypothalamic αMSH secretion is related to many metabolic diseases, such as obesity and type II diabetes. However, the underlying mechanisms are poorly understood. In our study, we focused on the reactive oxygen species (ROS)-induced adipocyte apoptosis and tried to unveil the role of αMSH in this process and the signal pathway which αMSH acts through. Kunming white mice were used and induced to oxidative stress status by hydrogen peroxide (H2O2) injection and a significant reduction of αMSH were found in mice serum, while elevated ROS level and mRNA level of pro-apoptotic genes were observed in mice adipose tissue. What is more, when detect the function of αMSH in ROS-induced apoptosis, similar inhibitory trend was found with the oxidative stress inhibitor N-acetyl-L-cysteine (NAC) in ROS-induced adipocyte apoptosis and this trend is αMSH receptor melanocortin 5 receptor (MC5R) depended, while an opposite trend was found between αMSH and Foxo1, which is a known positive regulator of adipocyte apoptosis. Further, we found that the repress effect of αMSH in adipocytes apoptosis is acting through Foxo1/mTORC2 pathway. These findings indicate that, αMSH has a strong inhibitory effect on ROS-induced adipocyte apoptosis and underlying mechanism is interacting with key factors in mTOR signal pathway. Our study demonstrated a great role of αMSH in adipocyte apoptosis and brings a new therapeutic mean to the treatment of obesity and diabetes.
Collapse
Affiliation(s)
- Weina Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Meihang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Tianjiao Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Fei Feng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Tongying Feng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yang Xu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
27
|
Yu F, Wei R, Yang J, Liu J, Yang K, Wang H, Mu Y, Hong T. FoxO1 inhibition promotes differentiation of human embryonic stem cells into insulin producing cells. Exp Cell Res 2017; 362:227-234. [PMID: 29157981 DOI: 10.1016/j.yexcr.2017.11.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 12/14/2022]
Abstract
Insulin-producing cells (IPCs) derived from human embryonic stem cells (hESCs) hold great potential for cell transplantation therapy in diabetes. Tremendous progress has been made in inducing differentiation of hESCs into IPCs in vitro, of which definitive endoderm (DE) protocol mimicking foetal pancreatic development has been widely used. However, immaturity of the obtained IPCs limits their further applications in treating diabetes. Forkhead box O1 (FoxO1) is involved in the differentiation and functional maintenance of murine pancreatic β cells, but its role in human β cell differentiation is under elucidation. Here, we showed that although FoxO1 expression level remained consistent, cytoplasmic phosphorylated FoxO1 protein level increased during IPC differentiation of hESCs induced by DE protocol. Lentiviral silencing of FoxO1 in pancreatic progenitors upregulated the levels of pancreatic islet differentiation-related genes and improved glucose-stimulated insulin secretion response in their progeny IPCs, whereas overexpression of FoxO1 showed the opposite effects. Notably, treatment with the FoxO1 inhibitor AS1842856 displayed similar effects with FoxO1 knockdown in pancreatic progenitors. These effects were closely associated with the mutually exclusive nucleocytoplasmic shuttling of FoxO1 and Pdx1 in the AS1842856-treated pancreatic progenitors. Our data demonstrated a promising effect of FoxO1 inhibition by the small molecule on gene expression profile during the differentiation, and in turn, on determining IPC maturation via modulating subcellular location of FoxO1 and Pdx1. Therefore, we identify a novel role of FoxO1 inhibition in promoting IPC differentiation of hESCs, which may provide clues for induction of mature β cells from hESCs and clinical applications in regenerative medicine.
Collapse
Affiliation(s)
- Fei Yu
- Department of Endocrinology and Metabolism, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Rui Wei
- Department of Endocrinology and Metabolism, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China.
| | - Jin Yang
- Department of Endocrinology and Metabolism, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Junling Liu
- Department of Endocrinology and Metabolism, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Kun Yang
- Department of Endocrinology and Metabolism, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Haining Wang
- Department of Endocrinology and Metabolism, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Yiming Mu
- Department of Endocrinology, Chinese PLA General Hospital, Beijing 100853, China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
28
|
Cheng CW, Villani V, Buono R, Wei M, Kumar S, Yilmaz OH, Cohen P, Sneddon JB, Perin L, Longo VD. Fasting-Mimicking Diet Promotes Ngn3-Driven β-Cell Regeneration to Reverse Diabetes. Cell 2017; 168:775-788.e12. [PMID: 28235195 PMCID: PMC5357144 DOI: 10.1016/j.cell.2017.01.040] [Citation(s) in RCA: 248] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/23/2016] [Accepted: 01/30/2017] [Indexed: 01/01/2023]
Abstract
Stem-cell-based therapies can potentially reverse organ dysfunction and diseases, but the removal of impaired tissue and activation of a program leading to organ regeneration pose major challenges. In mice, a 4-day fasting mimicking diet (FMD) induces a stepwise expression of Sox17 and Pdx-1, followed by Ngn3-driven generation of insulin-producing β cells, resembling that observed during pancreatic development. FMD cycles restore insulin secretion and glucose homeostasis in both type 2 and type 1 diabetes mouse models. In human type 1 diabetes pancreatic islets, fasting conditions reduce PKA and mTOR activity and induce Sox2 and Ngn3 expression and insulin production. The effects of the FMD are reversed by IGF-1 treatment and recapitulated by PKA and mTOR inhibition. These results indicate that a FMD promotes the reprogramming of pancreatic cells to restore insulin generation in islets from T1D patients and reverse both T1D and T2D phenotypes in mouse models. PAPERCLIP.
Collapse
Affiliation(s)
- Chia-Wei Cheng
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA; Koch Institute at MIT, 500 Main Street, Cambridge, MA 02139, USA
| | - Valentina Villani
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, University of Southern California, Los Angeles, Los Angeles, CA 90027, USA
| | - Roberta Buono
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA; IFOM FIRC Institute of Molecular Oncology, Via Adamello 16, Milan 20139, Italy
| | - Min Wei
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Sanjeev Kumar
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Omer H Yilmaz
- Koch Institute at MIT, 500 Main Street, Cambridge, MA 02139, USA
| | - Pinchas Cohen
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Julie B Sneddon
- Diabetes Center, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143
| | - Laura Perin
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, University of Southern California, Los Angeles, Los Angeles, CA 90027, USA
| | - Valter D Longo
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA; Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA; IFOM FIRC Institute of Molecular Oncology, Via Adamello 16, Milan 20139, Italy.
| |
Collapse
|
29
|
Jiang Z, Tian J, Zhang W, Yan H, Liu L, Huang Z, Lou J, Ma X. Forkhead Protein FoxO1 Acts as a Repressor to Inhibit Cell Differentiation in Human Fetal Pancreatic Progenitor Cells. J Diabetes Res 2017; 2017:6726901. [PMID: 28349071 PMCID: PMC5350409 DOI: 10.1155/2017/6726901] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/27/2016] [Accepted: 01/29/2017] [Indexed: 01/24/2023] Open
Abstract
Our colleagues have reported previously that human pancreatic progenitor cells can readily differentiate into insulin-containing cells. Particularly, transplantation of these cell clusters upon in vitro induction for 3-4 w partially restores hyperglycemia in diabetic nude mice. In this study, we used human fetal pancreatic progenitor cells to identify the forkhead protein FoxO1 as the key regulator for cell differentiation. Thus, induction of human fetal pancreatic progenitor cells for 1 week led to increase of the pancreatic β cell markers such as Ngn3, but decrease of stem cell markers including Oct4, Nanog, and CK19. Of note, FoxO1 knockdown or FoxO1 inhibitor significantly upregulated Ngn3 and insulin as well as the markers such as Glut2, Kir6.2, SUR1, and VDCC, which are designated for mature β cells. On the contrary, overexpression of FoxO1 suppressed the induction and reduced expression of these β cell markers. Taken together, these results suggest that FoxO1 may act as a repressor to inhibit cell differentiation in human fetal pancreatic progenitor cells.
Collapse
Affiliation(s)
- Zongzhe Jiang
- Shenzhen University Diabetes Institute, Shenzhen University, Shenzhen 518060, China
| | - Jingjing Tian
- Shenzhen University Diabetes Institute, Shenzhen University, Shenzhen 518060, China
| | - Wenjian Zhang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Hao Yan
- Shenzhen University Diabetes Institute, Shenzhen University, Shenzhen 518060, China
| | - Liping Liu
- Shenzhen Hightide Biopharmaceutical Ltd., Shenzhen 518000, China
| | - Zhenhe Huang
- Department of Aging Medicine, The Sixth Hospital of Shenzhen Municipality, Shenzhen 518060, China
| | - Jinning Lou
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xiaosong Ma
- Shenzhen University Diabetes Institute, Shenzhen University, Shenzhen 518060, China
- *Xiaosong Ma:
| |
Collapse
|
30
|
Aldehyde dehydrogenase 1a3 defines a subset of failing pancreatic β cells in diabetic mice. Nat Commun 2016; 7:12631. [PMID: 27572106 PMCID: PMC5013715 DOI: 10.1038/ncomms12631] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 07/18/2016] [Indexed: 12/21/2022] Open
Abstract
Insulin-producing β cells become dedifferentiated during diabetes progression. An impaired ability to select substrates for oxidative phosphorylation, or metabolic inflexibility, initiates progression from β-cell dysfunction to β-cell dedifferentiation. The identification of pathways involved in dedifferentiation may provide clues to its reversal. Here we isolate and functionally characterize failing β cells from various experimental models of diabetes and report a striking enrichment in the expression of aldehyde dehydrogenase 1 isoform A3 (ALDH(+)) as β cells become dedifferentiated. Flow-sorted ALDH(+) islet cells demonstrate impaired glucose-induced insulin secretion, are depleted of Foxo1 and MafA, and include a Neurogenin3-positive subset. RNA sequencing analysis demonstrates that ALDH(+) cells are characterized by: (i) impaired oxidative phosphorylation and mitochondrial complex I, IV and V; (ii) activated RICTOR; and (iii) progenitor cell markers. We propose that impaired mitochondrial function marks the progression from metabolic inflexibility to dedifferentiation in the natural history of β-cell failure.
Collapse
|
31
|
McCauley HA, Wells JM. Sweet Relief: Reprogramming Gastric Endocrine Cells to Make Insulin. Cell Stem Cell 2016; 18:295-7. [PMID: 26942844 DOI: 10.1016/j.stem.2016.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Two promising approaches toward a cell-based cure for diabetes are the directed differentiation of pluripotent stem cells or lineage reprogramming of other cell types to generate beta cells. In this issue of Cell Stem Cell, Ariyachet et al. (2016) reprogrammed gastric endocrine cells to generate a renewable source of insulin-producing cells.
Collapse
Affiliation(s)
- Heather A McCauley
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - James M Wells
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Endocrinology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.
| |
Collapse
|
32
|
Kim-Muller JY, Kim YJR, Fan J, Zhao S, Banks AS, Prentki M, Accili D. FoxO1 Deacetylation Decreases Fatty Acid Oxidation in β-Cells and Sustains Insulin Secretion in Diabetes. J Biol Chem 2016; 291:10162-72. [PMID: 26984405 DOI: 10.1074/jbc.m115.705608] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Indexed: 11/06/2022] Open
Abstract
Pancreatic β-cell dysfunction contributes to onset and progression of type 2 diabetes. In this state β-cells become metabolically inflexible, losing the ability to select between carbohydrates and lipids as substrates for mitochondrial oxidation. These changes lead to β-cell dedifferentiation. We have proposed that FoxO proteins are activated through deacetylation-dependent nuclear translocation to forestall the progression of these abnormalities. However, how deacetylated FoxO exert their actions remains unclear. To address this question, we analyzed islet function in mice homozygous for knock-in alleles encoding deacetylated FoxO1 (6KR). Islets expressing 6KR mutant FoxO1 have enhanced insulin secretion in vivo and ex vivo and decreased fatty acid oxidation ex vivo Remarkably, the gene expression signature associated with FoxO1 deacetylation differs from wild type by only ∼2% of the >4000 genes regulated in response to re-feeding. But this narrow swath includes key genes required for β-cell identity, lipid metabolism, and mitochondrial fatty acid and solute transport. The data support the notion that deacetylated FoxO1 protects β-cell function by limiting mitochondrial lipid utilization and raise the possibility that inhibition of fatty acid oxidation in β-cells is beneficial to diabetes treatment.
Collapse
Affiliation(s)
- Ja Young Kim-Muller
- From the Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, New York 10032, Merck Research Laboratories, Boston, Massachusetts 02816
| | - Young Jung R Kim
- From the Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, New York 10032
| | - Jason Fan
- From the Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, New York 10032
| | - Shangang Zhao
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the CRCHUM and Departments of Nutrition and Biochemistry and Department of Molecular Medicine, Université de Montréal, Montréal, H2X 0A9, Canada, and
| | | | - Marc Prentki
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the CRCHUM and Departments of Nutrition and Biochemistry and Department of Molecular Medicine, Université de Montréal, Montréal, H2X 0A9, Canada, and
| | - Domenico Accili
- From the Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, New York 10032,
| |
Collapse
|
33
|
Zhang T, Kim DH, Xiao X, Lee S, Gong Z, Muzumdar R, Calabuig-Navarro V, Yamauchi J, Harashima H, Wang R, Bottino R, Alvarez-Perez JC, Garcia-Ocaña A, Gittes G, Dong HH. FoxO1 Plays an Important Role in Regulating β-Cell Compensation for Insulin Resistance in Male Mice. Endocrinology 2016; 157:1055-70. [PMID: 26727107 PMCID: PMC4769368 DOI: 10.1210/en.2015-1852] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
β-Cell compensation is an essential mechanism by which β-cells increase insulin secretion for overcoming insulin resistance to maintain euglycemia in obesity. Failure of β-cells to compensate for insulin resistance contributes to insulin insufficiency and overt diabetes. To understand the mechanism of β-cell compensation, we characterized the role of forkhead box O1 (FoxO1) in β-cell compensation in mice under physiological and pathological conditions. FoxO1 is a key transcription factor that serves as a nutrient sensor for integrating insulin signaling to cell metabolism, growth, and proliferation. We showed that FoxO1 improved β-cell compensation via 3 distinct mechanisms by increasing β-cell mass, enhancing β-cell glucose sensing, and augmenting β-cell antioxidative function. These effects accounted for increased glucose-stimulated insulin secretion and enhanced glucose tolerance in β-cell-specific FoxO1-transgenic mice. When fed a high-fat diet, β-cell-specific FoxO1-transgenic mice were protected from developing fat-induced glucose disorder. This effect was attributable to increased β-cell mass and function. Furthermore, we showed that FoxO1 activity was up-regulated in islets, correlating with the induction of physiological β-cell compensation in high-fat-induced obese C57BL/6J mice. These data characterize FoxO1 as a pivotal factor for orchestrating physiological adaptation of β-cell mass and function to overnutrition and obesity.
Collapse
Affiliation(s)
- Ting Zhang
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Dae Hyun Kim
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Xiangwei Xiao
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Sojin Lee
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Zhenwei Gong
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Radhika Muzumdar
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Virtu Calabuig-Navarro
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Jun Yamauchi
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Hideyoshi Harashima
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Rennian Wang
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Rita Bottino
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Juan Carlos Alvarez-Perez
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - Adolfo Garcia-Ocaña
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - George Gittes
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| | - H Henry Dong
- Division of Pediatric Endocrinology (T.Z., D.H.K., S.L., Z.G., R.M., V.C.-N., J.Y., H.H.D.), Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Molecular Inflammation Research Center for Aging Intervention (D.H.K.), College of Pharmacy, Pusan National University, Busan, 609-735 Korea; Division of Pediatric Surgery (X.X., G.G.), Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Laboratory for Molecular Design of Pharmaceutics (J.Y., H.H.), Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812 Japan; Department of Physiology and Pharmacology (R.W.), University of Western Ontario, London, Ontario, N6C 2V5 Canada; Institute of Cellular Therapeutics (R.B.), Allegheny Health Network, Pittsburgh, Pennsylvania 15212; and Diabetes, Obesity and Metabolism Institute (J.C.A.-P., A.G.-O.), Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine Mt Sinai, New York, New York 10029
| |
Collapse
|
34
|
Kuo T, Kim-Muller JY, McGraw TE, Accili D. Altered Plasma Profile of Antioxidant Proteins as an Early Correlate of Pancreatic β Cell Dysfunction. J Biol Chem 2016; 291:9648-56. [PMID: 26917725 DOI: 10.1074/jbc.m115.702183] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Indexed: 12/22/2022] Open
Abstract
Insulin resistance and β cell dysfunction contribute to the pathogenesis of type 2 diabetes. Unlike insulin resistance, β cell dysfunction remains difficult to predict and monitor, because of the inaccessibility of the endocrine pancreas, the integrated relationship with insulin sensitivity, and the paracrine effects of incretins. The goal of our study was to survey the plasma response to a metabolic challenge in order to identify factors predictive of β cell dysfunction. To this end, we combined (i) the power of unbiased iTRAQ (isobaric tag for relative and absolute quantification) mass spectrometry with (ii) direct sampling of the portal vein following an intravenous glucose/arginine challenge (IVGATT) in (iii) mice with a genetic β cell defect. By so doing, we excluded the effects of peripheral insulin sensitivity as well as those of incretins on β cells, and focused on the first phase of insulin secretion to capture the early pathophysiology of β cell dysfunction. We compared plasma protein profiles with ex vivo islet secretome and transcriptome analyses. We detected changes to 418 plasma proteins in vivo, and detected changes to 262 proteins ex vivo The impairment of insulin secretion was associated with greater overall changes in the plasma response to IVGATT, possibly reflecting metabolic instability. Reduced levels of proteins regulating redox state and neuronal stress markers, as well as increased levels of coagulation factors, antedated the loss of insulin secretion in diabetic mice. These results suggest that a reduced complement of antioxidants in response to a mixed secretagogue challenge is an early correlate of future β cell failure.
Collapse
Affiliation(s)
- Taiyi Kuo
- From the Department of Medicine and Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, New York 10032 and
| | - Ja Young Kim-Muller
- From the Department of Medicine and Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, New York 10032 and
| | - Timothy E McGraw
- the Department of Biochemistry, Weill Cornell Medical College, New York, New York 10065
| | - Domenico Accili
- From the Department of Medicine and Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, New York 10032 and
| |
Collapse
|
35
|
Ariyachet C, Tovaglieri A, Xiang G, Lu J, Shah MS, Richmond CA, Verbeke C, Melton DA, Stanger BZ, Mooney D, Shivdasani RA, Mahony S, Xia Q, Breault DT, Zhou Q. Reprogrammed Stomach Tissue as a Renewable Source of Functional β Cells for Blood Glucose Regulation. Cell Stem Cell 2016; 18:410-21. [PMID: 26908146 DOI: 10.1016/j.stem.2016.01.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 10/05/2015] [Accepted: 01/08/2016] [Indexed: 12/12/2022]
Abstract
The gastrointestinal (GI) epithelium is a highly regenerative tissue with the potential to provide a renewable source of insulin(+) cells after undergoing cellular reprogramming. Here, we show that cells of the antral stomach have a previously unappreciated propensity for conversion into functional insulin-secreting cells. Native antral endocrine cells share a surprising degree of transcriptional similarity with pancreatic β cells, and expression of β cell reprogramming factors in vivo converts antral cells efficiently into insulin(+) cells with close molecular and functional similarity to β cells. Induced GI insulin(+) cells can suppress hyperglycemia in a diabetic mouse model for at least 6 months and regenerate rapidly after ablation. Reprogramming of antral stomach cells assembled into bioengineered mini-organs in vitro yielded transplantable units that also suppressed hyperglycemia in diabetic mice, highlighting the potential for development of engineered stomach tissues as a renewable source of functional β cells for glycemic control.
Collapse
Affiliation(s)
- Chaiyaboot Ariyachet
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Alessio Tovaglieri
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Guanjue Xiang
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Jiaqi Lu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, Peking University School of Pharmaceutical Sciences, Beijing 100191, China
| | - Manasvi S Shah
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Camilla A Richmond
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Catia Verbeke
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Douglas A Melton
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Ben Z Stanger
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David Mooney
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Ramesh A Shivdasani
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Shaun Mahony
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Qing Xia
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, Peking University School of Pharmaceutical Sciences, Beijing 100191, China
| | - David T Breault
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Qiao Zhou
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
36
|
Butler AE, Dhawan S, Hoang J, Cory M, Zeng K, Fritsch H, Meier JJ, Rizza RA, Butler PC. β-Cell Deficit in Obese Type 2 Diabetes, a Minor Role of β-Cell Dedifferentiation and Degranulation. J Clin Endocrinol Metab 2016; 101:523-32. [PMID: 26700560 PMCID: PMC4880126 DOI: 10.1210/jc.2015-3566] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
CONTEXT Type 2 diabetes is characterized by a β-cell deficit and a progressive defect in β-cell function. It has been proposed that the deficit in β-cells may be due to β-cell degranulation and transdifferentiation to other endocrine cell types. OBJECTIVE The objective of the study was to establish the potential impact of β-cell dedifferentiation and transdifferentiation on β-cell deficit in type 2 diabetes and to consider the alternative that cells with an incomplete identity may be newly forming rather than dedifferentiated. DESIGN, SETTING, AND PARTICIPANTS Pancreata obtained at autopsy were evaluated from 14 nondiabetic and 13 type 2 diabetic individuals, from four fetal cases, and from 10 neonatal cases. RESULTS Whereas there was a slight increase in islet endocrine cells expressing no hormone in type 2 diabetes (0.11 ± 0.03 cells/islet vs 0.03 ± 0.01 cells/islet, P < .01), the impact on the β-cell deficit would be minimal. Furthermore, we established that the deficit in β-cells per islet cannot be accounted for by an increase in other endocrine cell types. The distribution of hormone negative endocrine cells in type 2 diabetes (most abundant in cells scattered in the exocrine pancreas) mirrors that in developing (embryo and neonatal) pancreas, implying that these may represent newly forming cells. CONCLUSIONS Therefore, although we concur that in type 2 diabetes there are endocrine cells with altered cell identity, this process does not account for the deficit in β-cells in type 2 diabetes but may reflect, in part, attempted β-cell regeneration.
Collapse
Affiliation(s)
- Alexandra E Butler
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Sangeeta Dhawan
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Jonathan Hoang
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Megan Cory
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Kylie Zeng
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Helga Fritsch
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Juris J Meier
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Robert A Rizza
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Peter C Butler
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| |
Collapse
|
37
|
Gerst F, Kaiser G, Panse M, Sartorius T, Pujol A, Hennige AM, Machicao F, Lammers R, Bosch F, Häring HU, Ullrich S. Protein kinase Cδ regulates nuclear export of FOXO1 through phosphorylation of the chaperone 14-3-3ζ. Diabetologia 2015; 58:2819-31. [PMID: 26363783 DOI: 10.1007/s00125-015-3744-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/03/2015] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS Forkhead box protein O1 (FOXO1) is a transcription factor essential for beta cell fate. Protein kinase B-dependent phosphorylation of FOXO1 at S256 (P-FOXO1) enables its binding to 14-3-3 dimers and nuclear export. Dephosphorylated FOXO1 enters nuclei and activates pro-apoptotic genes. Since our previous observations suggest that protein kinase C delta (PKCδ) induces nuclear accumulation of FOXO1, the underlying mechanism was examined. METHODS In human islets, genetically modified mice and INS-1E cells apoptosis was assessed by TUNEL staining. Subcellular translocation of proteins was examined by confocal microscopy and signalling pathways were analysed by western blotting and overlay assay. RESULTS In PKCδ-overexpressing (PKCδ-tg) mouse islet cells and INS-1E cells FOXO1 accumulated in nuclei, surprisingly, as P-FOXO1. PKCδ-tg decelerated IGF-1-dependent stimulation of nuclear export, indicating that changes in export caused nuclear retention of P-FOXO1. Nuclear accumulation of P-FOXO1 was accompanied by increased phosphorylation of 14-3-3ζ at S58 and reduced dimerisation of 14-3-3ζ. Palmitic acid further augmented phosphorylation of 14-3-3ζ and triggered nuclear accumulation of FOXO1 in both INS-1E and human islet cells. Furthermore, the overexpression of a phosphomimicking mutant of 14-3-3ζ (S58D) enhanced nuclear FOXO1. In accordance with the nuclear accumulation of P-FOXO1, PKCδ overexpression alone did not increase apoptotic cell death. Additionally, insulin secretion and glucose homeostasis in PKCδ-overexpressing mice remained unaffected. CONCLUSIONS/INTERPRETATION These results suggest that PKCδ-mediated phosphorylation of 14-3-3ζ contributes to the nuclear retention of FOXO1, even when FOXO1 is phosphorylated as under non-stress conditions. P-FOXO1 does not induce pro-apoptotic genes, but may rather exert beneficial effects on beta cells.
Collapse
Affiliation(s)
- Felicia Gerst
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Gabriele Kaiser
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Madhura Panse
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
| | - Tina Sartorius
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Anna Pujol
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Anita M Hennige
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
| | - Fausto Machicao
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Reiner Lammers
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Hans-Ulrich Häring
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Susanne Ullrich
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany.
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany.
| |
Collapse
|