51
|
Tanday N, Irwin N, Flatt PR, Moffett RC. Dapagliflozin exerts positive effects on beta cells, decreases glucagon and does not alter beta- to alpha-cell transdifferentiation in mouse models of diabetes and insulin resistance. Biochem Pharmacol 2020; 177:114009. [PMID: 32360307 DOI: 10.1016/j.bcp.2020.114009] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 04/27/2020] [Indexed: 12/17/2022]
Abstract
Loss of beta cell identity and subsequent transdifferentiation of beta-to-alpha cells is implicated in the pathogenesis of diabetes. In addition, sodium-glucose transport protein 2 (SGLT2) inhibition has been linked to altered alpha-cell function. To investigate these phenomenon, lineage tracing of beta-cells was examined following 10-12 days dapagliflozin (1 or 5 mg/kg, once daily, as appropriate) treatment in multiple low-dose streptozotocin (STZ), high fat fed (HFF) or hydrocortisone (HC) transgenic Ins1Cre/+/Rosa26-eYFP mouse models of diabetes and insulin resistance. As anticipated, STZ, HFF and HC treated mice developed characteristic features of insulin deficiency or resistance. Dapagliflozin elicited differing beneficial effects depending on the aetiology of syndrome studied. The SGLT2 inhibitor efficiently promoted (P < 0.001) weight loss in HFF and STZ mice, whilst in HC mice it reduced (P < 0.001) energy intake, without an impact on body weight. Despite lacking significant effects on glycaemia, 1 mg/kg dapagliflozin consistently decreased both plasma and pancreatic glucagon. This was associated with increased pancreatic insulin in STZ and HFF mice. In STZ and HFF mice, beta cell proliferation and Pdx1 expression were enhanced by dapagliflozin, with a further increase in overall glucagon staining in HFF islets. Islet, beta- and alpha-cell areas were increased in dapagliflozin treated HC mice, which appeared to be linked to decreased alpha- and beta-cell apoptosis. Although the diabetes-like syndromes induced clear alterations in islet cell transdifferentiation, treatment with dapagliflozin (1 mg/kg) had no significant impact on this process, with 5 mg/kg marginally decreasing loss of beta-cells identity in STZ mice. These data suggest that SGLT2 inhibitors have positive effects on beta cells and decrease plasma and pancreatic glucagon, independent of changes in ambient glucose levels. Our combined data indicate that SGLT2 inhibitors do not directly induce hyperglucagonaemia.
Collapse
Affiliation(s)
- Neil Tanday
- From the SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - Nigel Irwin
- From the SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK.
| | - Peter R Flatt
- From the SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - R Charlotte Moffett
- From the SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| |
Collapse
|
52
|
Chawla P, Delgadillo Silva LF, Ninov N. Insights on β-cell regeneration from the zebrafish shoal: from generation of cells to functional integration. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2019.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
53
|
Thakur G, Lee HJ, Jeon RH, Lee SL, Rho GJ. Small Molecule-Induced Pancreatic β-Like Cell Development: Mechanistic Approaches and Available Strategies. Int J Mol Sci 2020; 21:E2388. [PMID: 32235681 PMCID: PMC7178115 DOI: 10.3390/ijms21072388] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/26/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes is a metabolic disease which affects not only glucose metabolism but also lipid and protein metabolism. It encompasses two major types: type 1 and 2 diabetes. Despite the different etiologies of type 1 and 2 diabetes mellitus (T1DM and T2DM, respectively), the defining features of the two forms are insulin deficiency and resistance, respectively. Stem cell therapy is an efficient method for the treatment of diabetes, which can be achieved by differentiating pancreatic β-like cells. The consistent generation of glucose-responsive insulin releasing cells remains challenging. In this review article, we present basic concepts of pancreatic organogenesis, which intermittently provides a basis for engineering differentiation procedures, mainly based on the use of small molecules. Small molecules are more auspicious than any other growth factors, as they have unique, valuable properties like cell-permeability, as well as a nonimmunogenic nature; furthermore, they offer immense benefits in terms of generating efficient functional beta-like cells. We also summarize advances in the generation of stem cell-derived pancreatic cell lineages, especially endocrine β-like cells or islet organoids. The successful induction of stem cells depends on the quantity and quality of available stem cells and the efficient use of small molecules.
Collapse
Affiliation(s)
- Gitika Thakur
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Hyeon-Jeong Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Ryoung-Hoon Jeon
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Sung-Lim Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Gyu-Jin Rho
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| |
Collapse
|
54
|
Sarnobat D, Moffett RC, Gault VA, Tanday N, Reimann F, Gribble FM, Flatt PR, Irwin N. Effects of long-acting GIP, xenin and oxyntomodulin peptide analogues on alpha-cell transdifferentiation in insulin-deficient diabetic Glu CreERT2;ROSA26-eYFP mice. Peptides 2020; 125:170205. [PMID: 31738969 PMCID: PMC7212078 DOI: 10.1016/j.peptides.2019.170205] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/08/2019] [Accepted: 11/13/2019] [Indexed: 02/08/2023]
Abstract
Enzyme-resistant long-acting forms of the gut-derived peptide hormones, glucose-dependent insulinotropic polypeptide (GIP), xenin and oxyntomodulin (Oxm) have been generated, and exert beneficial effects on diabetes control and pancreatic islet architecture. The current study has employed alpha-cell lineage tracing in GluCreERT2;ROSA26-eYFP transgenic mice to investigate the extent to which these positive pancreatic effects are associated with alpha- to beta-cell transdifferentiation. Twice-daily administration of (D-Ala2)GIP, xenin-25[Lys13PAL] or (D-Ser2)-Oxm[Lys38PAL] for 10 days to streptozotocin (STZ)-induced diabetic mice did not affect body weight, food intake or blood glucose levels, but (D-Ser2)-Oxm[Lys38PAL] reduced (P < 0.05 to P < 0.001) fluid intake and circulating glucagon. (D-Ala2)GIP and (D-Ser2)-Oxm[Lys38PAL] also augmented (P < 0.05 and P < 0.01, respectively) pancreatic insulin content. Detrimental changes of pancreatic morphology induced by STZ in GluCreERT2;ROSA26-eYFP mice were partially reversed by all treatment interventions. This was associated with reduced (P < 0.05) apoptosis and increased (P < 0.05 to P < 0.01) proliferation of beta-cells, alongside opposing effects on alpha-cells, with (D-Ala2)GIP and (D-Ser2)-Oxm[Lys38PAL] being particularly effective in this regard. Alpha-cell lineage tracing revealed that induction of diabetes was accompanied by increased (P < 0.01) transdifferentiation of glucagon positive alpha-cells to insulin positive beta-cells. This islet cell transitioning process was augmented (P < 0.01 and P < 0.001, respectively) by (D-Ala2)GIP and (D-Ser2)-Oxm[Lys38PAL]. (D-Ser2)-Oxm[Lys38PAL] also significantly (P < 0.05) promoted loss of alpha-cell identity in favour of other endocrine islet cells. These data highlight intra-islet benefits of (D-Ala2)GIP, xenin-25[Lys13PAL] and (D-Ser2)-Oxm[Lys38PAL] in diabetes with beta-cell loss induced by STZ. The effects appear to be independent of glycaemic change, and associated with alpha- to beta-cell transdifferentiation for the GIP and Oxm analogues.
Collapse
Affiliation(s)
- Dipak Sarnobat
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - R Charlotte Moffett
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - Victor A Gault
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - Neil Tanday
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - Frank Reimann
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Fiona M Gribble
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Peter R Flatt
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - Nigel Irwin
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, Northern Ireland, UK.
| |
Collapse
|
55
|
Yi Z, Waseem Ghani M, Ghani H, Jiang W, Waseem Birmani M, Ye L, Bin L, Cun LG, Lilong A, Mei X. Gimmicks of gamma-aminobutyric acid (GABA) in pancreatic β-cell regeneration through transdifferentiation of pancreatic α- to β-cells. Cell Biol Int 2020; 44:926-936. [PMID: 31903671 DOI: 10.1002/cbin.11302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 01/04/2020] [Indexed: 02/06/2023]
Abstract
In vivo regeneration of lost or dysfunctional islet β cells can fulfill the promise of improved therapy for diabetic patients. To achieve this, many mitogenic factors have been attempted, including gamma-aminobutyric acid (GABA). GABA remarkably affects pancreatic islet cells' (α cells and β cells) function through paracrine and/or autocrine binding to its membrane receptors on these cells. GABA has also been studied for promoting the transformation of α cells to β cells. Nonetheless, the gimmickry of GABA-induced α-cell transformation to β cells has two different perspectives. On the one hand, GABA was found to induce α-cell transformation to β cells in vivo and insulin-secreting β-like cells in vitro. On the other hand, GABA treatment showed that it has no α- to β-cell transformation response. Here, we will summarize the physiological effects of GABA on pancreatic islet β cells with an emphasis on its regenerative effects for transdifferentiation of islet α cells to β cells. We will also critically discuss the controversial results about GABA-mediated transdifferentiation of α cells to β cells.
Collapse
Affiliation(s)
- Zhao Yi
- Department of Animal Science and Medicine, Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China.,Department of Animal Breeding, Genetics and Reproduction, Agricultural Collage, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Muhammad Waseem Ghani
- Department of Animal Science and Medicine, Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China.,Department of Animal Breeding, Genetics and Reproduction, Agricultural Collage, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Hammad Ghani
- Nawaz Sharif Medical College, University of Gujrat, Punjab, 50180, Pakistan
| | - Wu Jiang
- Department of Animal Science and Medicine, Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China.,Department of Animal Breeding, Genetics and Reproduction, Agricultural Collage, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Muhammad Waseem Birmani
- Department of Animal Science and Medicine, Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Li Ye
- Department of Animal Science and Medicine, Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China.,Department of Animal Breeding, Genetics and Reproduction, Agricultural Collage, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Liu Bin
- Department of Animal Science and Medicine, Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China.,Department of Animal Breeding, Genetics and Reproduction, Agricultural Collage, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Lang Guan Cun
- Department of Animal Science and Medicine, Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China.,Department of Animal Breeding, Genetics and Reproduction, Agricultural Collage, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - An Lilong
- Department of Animal Science and Medicine, Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Xiao Mei
- Department of Animal Science and Medicine, Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China.,Department of Animal Breeding, Genetics and Reproduction, Agricultural Collage, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| |
Collapse
|
56
|
Ye L, Mueller O, Bagwell J, Bagnat M, Liddle RA, Rawls JF. High fat diet induces microbiota-dependent silencing of enteroendocrine cells. eLife 2019; 8:48479. [PMID: 31793875 PMCID: PMC6937151 DOI: 10.7554/elife.48479] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/26/2019] [Indexed: 12/18/2022] Open
Abstract
Enteroendocrine cells (EECs) are specialized sensory cells in the intestinal epithelium that sense and transduce nutrient information. Consumption of dietary fat contributes to metabolic disorders, but EEC adaptations to high fat feeding were unknown. Here, we established a new experimental system to directly investigate EEC activity in vivo using a zebrafish reporter of EEC calcium signaling. Our results reveal that high fat feeding alters EEC morphology and converts them into a nutrient insensitive state that is coupled to endoplasmic reticulum (ER) stress. We called this novel adaptation 'EEC silencing'. Gnotobiotic studies revealed that germ-free zebrafish are resistant to high fat diet induced EEC silencing. High fat feeding altered gut microbiota composition including enrichment of Acinetobacter bacteria, and we identified an Acinetobacter strain sufficient to induce EEC silencing. These results establish a new mechanism by which dietary fat and gut microbiota modulate EEC nutrient sensing and signaling.
Collapse
Affiliation(s)
- Lihua Ye
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, United States.,Division of Gastroenterology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Olaf Mueller
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, United States
| | - Jennifer Bagwell
- Department of Cell Biology, Duke University School of Medicine, Durham, United States
| | - Michel Bagnat
- Department of Cell Biology, Duke University School of Medicine, Durham, United States
| | - Rodger A Liddle
- Division of Gastroenterology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, United States.,Division of Gastroenterology, Department of Medicine, Duke University School of Medicine, Durham, United States
| |
Collapse
|
57
|
Mullapudi ST, Boezio GLM, Rossi A, Marass M, Matsuoka RL, Matsuda H, Helker CSM, Yang YHC, Stainier DYR. Disruption of the pancreatic vasculature in zebrafish affects islet architecture and function. Development 2019; 146:dev.173674. [PMID: 31597659 DOI: 10.1242/dev.173674] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 10/03/2019] [Indexed: 12/14/2022]
Abstract
A dense local vascular network is crucial for pancreatic endocrine cells to sense metabolites and secrete hormones, and understanding the interactions between the vasculature and the islets may allow for therapeutic modulation in disease conditions. Using live imaging in two models of vascular disruption in zebrafish, we identified two distinct roles for the pancreatic vasculature. At larval stages, expression of a dominant negative version of Vegfaa (dnVegfaa) in β-cells led to vascular and endocrine cell disruption with a minor impairment in β-cell function. In contrast, expression of a soluble isoform of Vegf receptor 1 (sFlt1) in β-cells blocked the formation of the pancreatic vasculature and drastically stunted glucose response, although islet architecture was not affected. Notably, these effects of dnVegfaa or sFlt1 were not observed in animals lacking vegfaa, vegfab, kdrl, kdr or flt1 function, indicating that they interfere with multiple ligands and/or receptors. In adults, disrupted islet architecture persisted in dnVegfaa-expressing animals, whereas sFlt1-expressing animals displayed large sheets of β-cells along their pancreatic ducts, accompanied by impaired glucose tolerance in both models. Thus, our study reveals novel roles for the vasculature in patterning and function of the islet.
Collapse
Affiliation(s)
- Sri Teja Mullapudi
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Giulia L M Boezio
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Andrea Rossi
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Michele Marass
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Ryota L Matsuoka
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Hiroki Matsuda
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Christian S M Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Yu Hsuan Carol Yang
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| |
Collapse
|
58
|
Houbrechts AM, Beckers A, Vancamp P, Sergeys J, Gysemans C, Mathieu C, Darras VM. Age-Dependent Changes in Glucose Homeostasis in Male Deiodinase Type 2 Knockout Zebrafish. Endocrinology 2019; 160:2759-2772. [PMID: 31504428 DOI: 10.1210/en.2019-00445] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 08/28/2019] [Indexed: 12/13/2022]
Abstract
Thyroid hormones (THs) are crucial regulators of glucose metabolism and insulin sensitivity. Moreover, inactivating mutations in type 2 deiodinase (DIO2), the major TH-activating enzyme, have been associated with type 2 diabetes mellitus in both humans and mice. We studied the link between Dio2 deficiency and glucose homeostasis in fasted males of two different Dio2 knockout (KO) zebrafish lines. Young adult Dio2KO zebrafish (6 to 9 months) were hyperglycemic. Both insulin and glucagon expression were increased, whereas β and α cell numbers in the main pancreatic islet were similar to those in wild-types. Insulin receptor expression in skeletal muscle was decreased at 6 months, accompanied by a strong downregulation of hexokinase and pyruvate kinase expression. Blood glucose levels in Dio2KO zebrafish, however, normalized around 1 year of age. Older mutants (18 to 24 months) were normoglycemic, and increased insulin and glucagon expression was accompanied by a prominent increase in pancreatic islet size and β and α cell numbers. Older Dio2KO zebrafish also showed strongly decreased expression of glucagon receptors in the gastrointestinal system as well as decreased expression of glucose transporters GLUT2 and GLUT12, glucose-6-phosphatase, and glycogen synthase 2. This study shows that Dio2KO zebrafish suffer from transient hyperglycemia, which is counteracted with increasing age by a prominent hyperplasia of the endocrine pancreas together with decreases in hepatic glucagon sensitivity and intestinal glucose uptake. Further research on the mechanisms allowing compensation in older Dio2KO zebrafish may help to identify new therapeutic targets for (TH deficiency-related) hyperglycemia.
Collapse
Affiliation(s)
- Anne M Houbrechts
- Laboratory of Comparative Endocrinology, Division of Animal Physiology and Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| | - An Beckers
- Laboratory of Neural Circuit Development and Regeneration, Division of Animal Physiology and Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Pieter Vancamp
- Laboratory of Comparative Endocrinology, Division of Animal Physiology and Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Jurgen Sergeys
- Laboratory of Neural Circuit Development and Regeneration, Division of Animal Physiology and Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Conny Gysemans
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism, and Aging, KU Leuven, Leuven, Belgium
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism, and Aging, KU Leuven, Leuven, Belgium
| | - Veerle M Darras
- Laboratory of Comparative Endocrinology, Division of Animal Physiology and Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| |
Collapse
|
59
|
Wang Y, Sun J, Ni Q, Nie A, Gu Y, Wang S, Zhang W, Ning G, Wang W, Wang Q. Dual Effect of Raptor on Neonatal β-Cell Proliferation and Identity Maintenance. Diabetes 2019; 68:1950-1964. [PMID: 31345937 DOI: 10.2337/db19-0166] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 07/11/2019] [Indexed: 11/13/2022]
Abstract
Immature pancreatic β-cells are highly proliferative, and the expansion of β-cells during the early neonatal period largely determines functional β-cell mass; however, the mechanisms are poorly characterized. We generated Ngn3RapKO mice (ablation of Raptor, an essential component of mechanistic target of rapamycin [mTORC1] in Ngn3+ endocrine progenitor cells) and found that mTORC1 was dispensable for endocrine cell lineage formation but specifically regulated both proliferation and identity maintenance of neonatal β-cells. Ablation of Raptor in neonatal β-cells led to autonomous loss of cell identity, decelerated cell cycle progression, compromised proliferation, and caused neonatal diabetes as a result of inadequate establishment of functional β-cell mass at postnatal day 14. Completely different from mature β-cells, Raptor regulated G1/S and G2/M phase cell cycle transition, thus permitting a high proliferation rate in neonatal β-cells. Moreover, Ezh2 was identified as a critical downstream target of mTORC1 in neonatal β-cells, which was responsible for G2/M phase transition and proliferation. Our discovery of the dual effect of mTORC1 in immature β-cells has revealed a potential target for replenishing functional β-cell pools by promoting both expansion and functional maturation of newly formed immature β-cells.
Collapse
Affiliation(s)
- Yanqiu Wang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiajun Sun
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qicheng Ni
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aifang Nie
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanyun Gu
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu Wang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Science, Peking University Health Science Center, Beijing, China
| | - Guang Ning
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qidi Wang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
60
|
Zeng Z, Yuan Q, Yu R, Zhang J, Ma H, Chen S. Ameliorative Effects of Probiotic Lactobacillus paracasei NL41 on Insulin Sensitivity, Oxidative Stress, and Beta-Cell Function in a Type 2 Diabetes Mellitus Rat Model. Mol Nutr Food Res 2019; 63:e1900457. [PMID: 31433912 DOI: 10.1002/mnfr.201900457] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/17/2019] [Indexed: 01/31/2023]
Abstract
SCOPE The present study aims to assess the antidiabetic effect of Lactobacillus paracasei strain NL41 and its potential mechanisms in rats with type 2 diabetes mellitus (T2DM) induced by a high-fat diet and low-dose streptozotocin administration (HFD/STZ). METHODS AND RESULTS Eighteen Sprague-Dawley (SD) rats are randomly assigned to three groups: one control, one HFD/STZ model, and one HFD/STZ-Lactobacillus protection group with administration of strain NL41 for 12 weeks. Blood is collected for biochemical parameters analysis and tissue samples for histological analysis. Treatment with strain NL41 results in excellent blood glucose regulation and significantly decreases insulin resistance, and HbA1c, glucagon, and leptin levels, accompanied by remarkable improvement of dyslipidemia and oxidative stress status in the animals. Islets of Langerhans, liver, and kidney are significantly protected in the NL41-treated rats compared to the HFD/STZ-T2DM model rats. Histochemistry shows that strain NL41 inhibits beta-cell loss and alpha-cell expansion, indicating pancreatic islets as the targeted tissues for the primary ameliorative effect of the probiotic strain on HFD/STZ-T2DM rats. Crosstalk between the gut-liver and liver-pancreas endocrine axes is discussed. CONCLUSION Probiotic strain NL41 prevents HFD/STZ-T2DM by decreasing insulin resistance and oxidative stress status, and protecting beta-cell function.
Collapse
Affiliation(s)
- Zhu Zeng
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Biotechnology, Southwest University, Chongqing, 400715, P. R. China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P. R. China
| | - Qipeng Yuan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P. R. China
| | - Rui Yu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P. R. China
| | - Jinlan Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P. R. China
| | - Huiqin Ma
- College of Horticulture, China Agricultural University, Beijing, 100193, P. R. China
| | - Shangwu Chen
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P. R. China
| |
Collapse
|
61
|
Abstract
PURPOSE OF REVIEW This review summarizes the alterations in the β-cell observed in type 2 diabetes (T2D), focusing on changes in β-cell identity and mass and changes associated with metabolism and intracellular signaling. RECENT FINDINGS In the setting of T2D, β-cells undergo changes in gene expression, reverting to a more immature state and in some cases transdifferentiating into other islet cell types. Alleviation of metabolic stress, ER stress, and maladaptive prostaglandin signaling could improve β-cell function and survival. The β-cell defects leading to T2D likely differ in different individuals and include variations in β-cell mass, development, β-cell expansion, responses to ER and oxidative stress, insulin production and secretion, and intracellular signaling pathways. The recent recognition that some β-cells undergo dedifferentiation without dying in T2D suggests strategies to revive these cells and rejuvenate their functionality.
Collapse
Affiliation(s)
- Ashley A Christensen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Maureen Gannon
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA.
- Department of Medicine, Vanderbilt University Medical Center, 2213 Garland Ave, MRB IV 7465, Nashville, TN, 37232, USA.
- Department of Veterans Affairs, Tennessee Valley Health Authority, Nashville, TN, 37232, USA.
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
62
|
Helker CSM, Mullapudi ST, Mueller LM, Preussner J, Tunaru S, Skog O, Kwon HB, Kreuder F, Lancman JJ, Bonnavion R, Dong PDS, Looso M, Offermanns S, Korsgren O, Spagnoli FM, Stainier DYR. A whole organism small molecule screen identifies novel regulators of pancreatic endocrine development. Development 2019; 146:dev.172569. [PMID: 31142539 DOI: 10.1242/dev.172569] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 05/08/2019] [Indexed: 12/16/2022]
Abstract
An early step in pancreas development is marked by the expression of the transcription factor Pdx1 within the pancreatic endoderm, where it is required for the specification of all endocrine cell types. Subsequently, Pdx1 expression becomes restricted to the β-cell lineage, where it plays a central role in β-cell function. This pivotal role of Pdx1 at various stages of pancreas development makes it an attractive target to enhance pancreatic β-cell differentiation and increase β-cell function. In this study, we used a newly generated zebrafish reporter to screen over 8000 small molecules for modulators of pdx1 expression. We found four hit compounds and validated their efficacy at different stages of pancreas development. Notably, valproic acid treatment increased pancreatic endoderm formation, while inhibition of TGFβ signaling led to α-cell to β-cell transdifferentiation. HC toxin, another HDAC inhibitor, enhances β-cell function in primary mouse and human islets. Thus, using a whole organism screening strategy, this study identified new pdx1 expression modulators that can be used to influence different steps in pancreas and β-cell development.
Collapse
Affiliation(s)
- Christian S M Helker
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, 61231 Bad Nauheim, Germany .,Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, 35043 Marburg, Germany
| | - Sri-Teja Mullapudi
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, 61231 Bad Nauheim, Germany
| | - Laura M Mueller
- Centre for Stem Cells and Regenerative Medicine, King's College London, London WC2R 2LS, UK
| | - Jens Preussner
- Max Planck Institute for Heart and Lung Research, ECCPS Bioinformatics Core Unit, 61231 Bad Nauheim, Germany
| | - Sorin Tunaru
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, 61231 Bad Nauheim, Germany.,Biochemistry Institute of the Romanian Academy, Department of Enzymology, Bucharest 060031, Romania
| | - Oskar Skog
- Uppsala University, Department of Immunology, Genetics and Pathology, 751 85 Uppsala, Sweden
| | - Hyouk-Bum Kwon
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, 61231 Bad Nauheim, Germany
| | - Florian Kreuder
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, 61231 Bad Nauheim, Germany
| | - Joseph J Lancman
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.,Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Remy Bonnavion
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, 61231 Bad Nauheim, Germany
| | - P Duc Si Dong
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.,Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Mario Looso
- Max Planck Institute for Heart and Lung Research, ECCPS Bioinformatics Core Unit, 61231 Bad Nauheim, Germany
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, 61231 Bad Nauheim, Germany
| | - Ole Korsgren
- Uppsala University, Department of Immunology, Genetics and Pathology, 751 85 Uppsala, Sweden
| | - Francesca M Spagnoli
- Centre for Stem Cells and Regenerative Medicine, King's College London, London WC2R 2LS, UK
| | - Didier Y R Stainier
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, 61231 Bad Nauheim, Germany
| |
Collapse
|
63
|
Szlapinski SK, King RT, Retta G, Yeo E, Strutt BJ, Hill DJ. A mouse model of gestational glucose intolerance through exposure to a low protein diet during fetal and neonatal development. J Physiol 2019; 597:4237-4250. [PMID: 31206692 DOI: 10.1113/jp277884] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/10/2019] [Indexed: 12/25/2022] Open
Abstract
KEY POINTS Pancreatic β-cell dysfunction is hypothesized to be the significant determinant of gestational diabetes pathogenesis, however pancreatic samples from patients are scarce. This study reports a novel mouse model of gestational glucose intolerance in pregnancy, originating from previous nutrition restriction in utero, in which glucose intolerance was restricted to late gestation as is seen in human gestational diabetes. Glucose intolerance was attributed to reduced β-cell proliferation, leading to impaired gestational β-cell mass expansion in maternal endocrine pancreas, in addition to reduced glucose-stimulated insulin secretion. This model reproduces some of the features of gestational diabetes and is suitable for testing safe therapeutic interventions that increase β-cell mass during pregnancy and prevent or reverse gestational glucose intolerance. ABSTRACT Gestational diabetes mellitus (GDM) is an increasingly prevalent form of diabetes that appears during pregnancy. Pathological studies link a failure to adaptively increase maternal pancreatic β-cell mass (BCM) in pregnancy to GDM. Due to the scarcity of pancreatic samples from GDM patients, we sought to develop a novel mouse model for impaired gestational glucose tolerance. Mature female C57Bl/6 mouse offspring (F1) born to dams fed either a control (C) or low-protein (LP) diet during gestation and lactation were randomly allocated into two subsequent study groups: pregnant (CP, LPP) or non-pregnant (CNP, LPNP). Glucose tolerance tests were performed at gestational day (GD) 9, 12 and 18. Subsequently, pancreata were removed for fluorescence immunohistochemistry to assess α-cell mass (ACM), BCM and β-cell proliferation. An additional group of animals was used to measure insulin secretion from isolated islets at GD18. LPP females displayed glucose intolerance compared to CP females at GD18 (P < 0.001). BCM increased threefold at GD18 in CP females. However, LPP females had reduced BCM expansion (P < 0.01) concurrent with reduced β-cell proliferation at GD12 (P < 0.05). LPP females also had reduced ACM expansion at GD18 (P < 0.01). LPP islets had impaired glucose-stimulated insulin secretion in vitro compared to CP islets (P < 0.01). Therefore, impaired glucose tolerance during pregnancy is associated with a failure to adequately adapt BCM, as a result of reduced β-cell proliferation, in addition to lower glucose-stimulated insulin secretion. This model could be used to evaluate novel interventions during pregnancy to increase BCM or function as a strategy to prevent/reverse GDM.
Collapse
Affiliation(s)
- Sandra K Szlapinski
- Department of Physiology and Pharmacology, Western University, 1151 Richmond St., London, ON, Canada.,Lawson Health Research Institute, St Joseph's Health Care, 268 Grosvenor St., F4-124, London, ON, Canada
| | - Renee T King
- Lawson Health Research Institute, St Joseph's Health Care, 268 Grosvenor St., F4-124, London, ON, Canada
| | - Gabrielle Retta
- Lawson Health Research Institute, St Joseph's Health Care, 268 Grosvenor St., F4-124, London, ON, Canada
| | - Erica Yeo
- Lawson Health Research Institute, St Joseph's Health Care, 268 Grosvenor St., F4-124, London, ON, Canada
| | - Brenda J Strutt
- Department of Physiology and Pharmacology, Western University, 1151 Richmond St., London, ON, Canada.,Lawson Health Research Institute, St Joseph's Health Care, 268 Grosvenor St., F4-124, London, ON, Canada
| | - David J Hill
- Department of Physiology and Pharmacology, Western University, 1151 Richmond St., London, ON, Canada.,Lawson Health Research Institute, St Joseph's Health Care, 268 Grosvenor St., F4-124, London, ON, Canada
| |
Collapse
|
64
|
Chen J, He J, Ni R, Yang Q, Zhang Y, Luo L. Cerebrovascular Injuries Induce Lymphatic Invasion into Brain Parenchyma to Guide Vascular Regeneration in Zebrafish. Dev Cell 2019; 49:697-710.e5. [PMID: 31006646 DOI: 10.1016/j.devcel.2019.03.022] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 02/11/2019] [Accepted: 03/22/2019] [Indexed: 12/21/2022]
Abstract
Damage to regional cerebrovascular networks and neuronal tissues occurs during acute cerebrovascular diseases, such as ischemic stroke. The promotion of vascular regeneration is the most promising therapeutic approach. To understand the cellular and molecular mechanisms underlying brain vascular regeneration, we developed two zebrafish cerebrovascular injury models using genetic ablation and photochemical thrombosis. Although brain parenchyma is physiologically devoid of lymphatic vasculature, we found that cerebrovascular injuries induce rapid ingrowth of meningeal lymphatics into the injured parenchyma. The ingrown lymphatics on one hand become lumenized to drain interstitial fluid to resolve brain edema and on the other hand act as "growing tracks" for nascent blood vessels. The ingrown lymphatic vessels undergo apoptosis and clearance after cerebrovascular regeneration. This study reveals a pathological function of meningeal lymphatics, through previously unexpected ingrowth into brain parenchyma and a newly identified lymphatic function as vascular "growing tracks."
Collapse
Affiliation(s)
- Jingying Chen
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Jianbo He
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Rui Ni
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Qifen Yang
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Yaoguang Zhang
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Lingfei Luo
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China.
| |
Collapse
|
65
|
Naylor RW, Chang HHG, Qubisi S, Davidson AJ. A novel mechanism of gland formation in zebrafish involving transdifferentiation of renal epithelial cells and live cell extrusion. eLife 2018; 7:38911. [PMID: 30394875 PMCID: PMC6250424 DOI: 10.7554/elife.38911] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 11/05/2018] [Indexed: 12/12/2022] Open
Abstract
Transdifferentiation is the poorly understood phenomenon whereby a terminally differentiated cell acquires a completely new identity. Here, we describe a rare example of a naturally occurring transdifferentiation event in zebrafish in which kidney distal tubule epithelial cells are converted into an endocrine gland known as the Corpuscles of Stannius (CS). We find that this process requires Notch signalling and is associated with the cytoplasmic sequestration of the Hnf1b transcription factor, a master-regulator of renal tubule fate. A deficiency in the Irx3b transcription factor results in ectopic transdifferentiation of distal tubule cells to a CS identity but in a Notch-dependent fashion. Using live-cell imaging we show that CS cells undergo apical constriction en masse and are then extruded from the tubule to form a distinct organ. This system provides a valuable new model to understand the molecular and morphological basis of transdifferentiation and will advance efforts to exploit this rare phenomenon therapeutically.
Collapse
Affiliation(s)
- Richard W Naylor
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Hao-Han G Chang
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Sarah Qubisi
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Alan J Davidson
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
66
|
Liu P, Song J, Liu H, Yan F, He T, Wang L, Shen H, Hou X, Chen L. Insulin regulates glucagon-like peptide-1 secretion by pancreatic alpha cells. Endocrine 2018; 62:394-403. [PMID: 30084102 DOI: 10.1007/s12020-018-1684-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 06/05/2018] [Indexed: 12/19/2022]
Abstract
PURPOSE Proglucagon is expressed in both pancreatic alpha cells and intestinal epithelial L cells and is cleaved into glucagon and glucagon-like peptide-1 (GLP-1) by different prohormone convertases (PCs). Recent studies have shown that α-cells can also secrete GLP-1, which may improve islet function. However, little is known about the factors influencing GLP-1 secretion by α cells. In this study, we investigated whether insulin promotes GLP-1 secretion by α cells, as well as the mechanisms underlying this phenomenon. METHODS We cultured the alpha-cell line In-R1-G9 in low- or high-glucose medium in the presence or absence of insulin to determine the influence of glucose concentrations on the actions of insulin. We also treated In-R1-G9 cells with insulin for different times and at different doses. Then GLP-1 and glucagon protein expression levels were estimated. Moreover, ERK and phosphatidylinositol-3-kinase/AKT (PI3K/AKT) pathway activity levels and prohormone convertase expression levels were evaluated to elucidate the mechanism underlying the effects of insulin on GLP-1 secretion by α-cells. RESULTS Insulin promoted GLP-1 secretion in a time- and dose-dependent manner under high-glucose conditions. Inhibiting the PI3K/AKT pathway with LY294002 and the Ras/mitogen-activated protein kinase (RAS/MAPK) pathway with PD98059 reduced GLP-1 secretion, respectively, in inhibitor-treated cells compared with insulin-treated cells. Moreover, insulin increased prohormone convertase 1/3 expression levels in the corresponding group of IN-R1-G9 cells compared with the control group of cells. CONCLUSION Insulin facilitates GLP-1 secretion by pancreatic alpha cells by inducing PC1/3 expression under high-glucose conditions, a phenomenon that may be associated mainly with PI3K/AKT pathway activation.
Collapse
Affiliation(s)
- Pan Liu
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250000, China
- Institute of Endocrinology and Metabolism, Shandong University, Jinan, 250000, China
- Key Laboratory of Endocrinology and Metabolism, Shandong Province in Medicine & Health, Jinan, 250000, China
| | - Jia Song
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250000, China
- Institute of Endocrinology and Metabolism, Shandong University, Jinan, 250000, China
- Key Laboratory of Endocrinology and Metabolism, Shandong Province in Medicine & Health, Jinan, 250000, China
| | - He Liu
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250000, China
- Institute of Endocrinology and Metabolism, Shandong University, Jinan, 250000, China
- Key Laboratory of Endocrinology and Metabolism, Shandong Province in Medicine & Health, Jinan, 250000, China
| | - Fei Yan
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250000, China
- Institute of Endocrinology and Metabolism, Shandong University, Jinan, 250000, China
- Key Laboratory of Endocrinology and Metabolism, Shandong Province in Medicine & Health, Jinan, 250000, China
| | - Tianyi He
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250000, China
- Institute of Endocrinology and Metabolism, Shandong University, Jinan, 250000, China
- Key Laboratory of Endocrinology and Metabolism, Shandong Province in Medicine & Health, Jinan, 250000, China
| | - Lingshu Wang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250000, China
- Institute of Endocrinology and Metabolism, Shandong University, Jinan, 250000, China
- Key Laboratory of Endocrinology and Metabolism, Shandong Province in Medicine & Health, Jinan, 250000, China
| | - Huying Shen
- Baylor College of Medicine, Houston, TX, USA
| | - Xinguo Hou
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250000, China.
- Institute of Endocrinology and Metabolism, Shandong University, Jinan, 250000, China.
- Key Laboratory of Endocrinology and Metabolism, Shandong Province in Medicine & Health, Jinan, 250000, China.
| | - Li Chen
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250000, China.
- Institute of Endocrinology and Metabolism, Shandong University, Jinan, 250000, China.
- Key Laboratory of Endocrinology and Metabolism, Shandong Province in Medicine & Health, Jinan, 250000, China.
| |
Collapse
|
67
|
Xu J, Jia YF, Tapadar S, Weaver JD, Raji IO, Pithadia DJ, Javeed N, García AJ, Choi DS, Matveyenko AV, Oyelere AK, Shin CH. Inhibition of TBK1/IKKε Promotes Regeneration of Pancreatic β-cells. Sci Rep 2018; 8:15587. [PMID: 30349097 PMCID: PMC6197228 DOI: 10.1038/s41598-018-33875-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/01/2018] [Indexed: 12/18/2022] Open
Abstract
β-cell proliferation induction is a promising therapeutic strategy to restore β-cell mass. By screening small molecules in a transgenic zebrafish model of type 1 diabetes, we identified inhibitors of non-canonical IκB kinases (IKKs), TANK-binding kinase 1 (TBK1) and IκB kinase ε (IKKε), as enhancers of β-cell regeneration. The most potent β-cell regeneration enhancer was a cinnamic acid derivative (E)-3-(3-phenylbenzo[c]isoxazol-5-yl)acrylic acid (PIAA), which, acting through the cAMP-dependent protein kinase A (PKA), stimulated β-cell-specific proliferation by increasing cyclic AMP (cAMP) levels and mechanistic target of rapamycin (mTOR) activity. A combination of PIAA and cilostamide, an inhibitor of β-cell-enriched cAMP hydrolyzing enzyme phosphodiesterase (PDE) 3, enhanced β-cell proliferation, whereas overexpression of PDE3 blunted the mitogenic effect of PIAA in zebrafish. PIAA augmented proliferation of INS-1β-cells and β-cells in mammalian islets including human islets with elevation in cAMP levels and insulin secretion. PIAA improved glycemic control in streptozotocin (STZ)-induced diabetic mice with increases in β-cell proliferation, β-cell area, and insulin content in the pancreas. Collectively, these data reveal an evolutionarily conserved and critical role of TBK1/IKKε suppression in expanding functional β-cell mass.
Collapse
Affiliation(s)
- Jin Xu
- School of Biological Sciences and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Yun-Fang Jia
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Subhasish Tapadar
- School of Chemistry and Biochemistry and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jessica D Weaver
- Woodruff School of Mechanical Engineering and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Idris O Raji
- School of Chemistry and Biochemistry and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Deeti J Pithadia
- School of Biological Sciences and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Naureen Javeed
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Aleksey V Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Adegboyega K Oyelere
- School of Chemistry and Biochemistry and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Chong Hyun Shin
- School of Biological Sciences and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA. .,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
68
|
Earley AM, Graves CL, Shiau CE. Critical Role for a Subset of Intestinal Macrophages in Shaping Gut Microbiota in Adult Zebrafish. Cell Rep 2018; 25:424-436. [PMID: 30304682 PMCID: PMC6245655 DOI: 10.1016/j.celrep.2018.09.025] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 06/27/2018] [Accepted: 09/07/2018] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota is strongly influenced by environmental factors, although host contribution is far less understood. We leveraged macrophage-deficient interferon regulatory factor irf8 zebrafish mutants to investigate the role of macrophages in this process. In conventionally raised adult irf8-deficient mutants, we found a significant loss of intestinal macrophages associated with a strikingly altered gut microbiota when compared to co-housed siblings. The destabilization of the gut commensal microbiota was associated with a severe reduction in complement C1q genes and outgrowth of a rare bacterial species. Consistent with a critical function of irf8 in adult intestinal macrophages, irf8 is abundantly expressed in these cells normally, and restoring macrophage irf8 expression in irf8 mutants was sufficient to recover commensal microbes and C1q genes expression. This study reports an important subpopulation of intestinal macrophages that requires irf8 to establish in the gut, ensure normal colonization of gut microbes, and prevent immune dysregulation.
Collapse
Affiliation(s)
- Alison M Earley
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christina L Graves
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Celia E Shiau
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
69
|
Zang L, Maddison LA, Chen W. Zebrafish as a Model for Obesity and Diabetes. Front Cell Dev Biol 2018; 6:91. [PMID: 30177968 PMCID: PMC6110173 DOI: 10.3389/fcell.2018.00091] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/25/2018] [Indexed: 12/13/2022] Open
Abstract
Obesity and diabetes now considered global epidemics. The prevalence rates of diabetes are increasing in parallel with the rates of obesity and the strong connection between these two diseases has been coined as “diabesity.” The health risks of overweight or obesity include Type 2 diabetes mellitus (T2DM), coronary heart disease and cancer of numerous organs. Both obesity and diabetes are complex diseases that involve the interaction of genetics and environmental factors. The underlying pathogenesis of obesity and diabetes are not well understood and further research is needed for pharmacological and surgical management. Consequently, the use of animal models of obesity and/or diabetes is important for both improving the understanding of these diseases and to identify and develop effective treatments. Zebrafish is an attractive model system for studying metabolic diseases because of the functional conservation in lipid metabolism, adipose biology, pancreas structure, and glucose homeostasis. It is also suited for identification of novel targets associated with the risk and treatment of obesity and diabetes in humans. In this review, we highlight studies using zebrafish to model metabolic diseases, and discuss the advantages and disadvantages of studying pathologies associated with obesity and diabetes in zebrafish.
Collapse
Affiliation(s)
- Liqing Zang
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States.,Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| | - Lisette A Maddison
- Center for Reproductive Biology, Washington State University, Pullman, WA, United States
| | - Wenbiao Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
70
|
Matsuda H. Zebrafish as a model for studying functional pancreatic β cells development and regeneration. Dev Growth Differ 2018; 60:393-399. [DOI: 10.1111/dgd.12565] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/06/2018] [Accepted: 07/06/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Hiroki Matsuda
- Department of Developmental Biology and Neurosciences; Graduate School of Life Sciences; Tohoku University; Sendai Japan
- Department of Biomedical Sciences; College of Life Sciences; Ritsumeikan University; Kusatsu Japan
| |
Collapse
|
71
|
Karanth S, Adams JD, Serrano MDLA, Quittner-Strom EB, Simcox J, Villanueva CJ, Ozcan L, Holland WL, Yost HJ, Vella A, Schlegel A. A Hepatocyte FOXN3-α Cell Glucagon Axis Regulates Fasting Glucose. Cell Rep 2018; 24:312-319. [PMID: 29996093 DOI: 10.1016/j.celrep.2018.06.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/07/2018] [Accepted: 06/08/2018] [Indexed: 01/26/2023] Open
Abstract
The common genetic variation at rs8004664 in the FOXN3 gene is independently and significantly associated with fasting blood glucose, but not insulin, in non-diabetic humans. Recently, we reported that primary hepatocytes from rs8004664 hyperglycemia risk allele carriers have increased FOXN3 transcript and protein levels and liver-limited overexpression of human FOXN3, a transcriptional repressor that had not been implicated in metabolic regulation previously, increases fasting blood glucose in zebrafish. Here, we find that injection of glucagon into mice and adult zebrafish decreases liver Foxn3 protein and transcript levels. Zebrafish foxn3 loss-of-function mutants have decreased fasting blood glucose, blood glucagon, liver gluconeogenic gene expression, and α cell mass. Conversely, liver-limited overexpression of foxn3 increases α cell mass. Supporting these genetic findings in model organisms, non-diabetic rs8004664 risk allele carriers have decreased suppression of glucagon during oral glucose tolerance testing. By reciprocally regulating each other, liver FOXN3 and glucagon control fasting glucose.
Collapse
Affiliation(s)
- Santhosh Karanth
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Internal Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - J D Adams
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Maria de Los Angeles Serrano
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Ezekiel B Quittner-Strom
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA
| | - Judith Simcox
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Claudio J Villanueva
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Lale Ozcan
- Department of Medicine, Division of Molecular Medicine, Columbia University Medical Center, New York, NY, USA
| | - William L Holland
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA; Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - H Joseph Yost
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Adrian Vella
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Amnon Schlegel
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Internal Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA; Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA.
| |
Collapse
|
72
|
Webb MA, Chen JJ, James RFL, Davies MJ, Dennison AR. Elevated Levels of Alpha Cells Emanating from the Pancreatic Ducts of a Patient with a Low BMI and Chronic Pancreatitis. Cell Transplant 2018; 27:902-906. [PMID: 29852747 PMCID: PMC6050909 DOI: 10.1177/0963689718755707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chronic pancreatitis (CP) is an inflammatory disease that causes progressive damage to
the pancreatic parenchyma with irreversible morphological changes and fibrotic replacement
of the gland. The risk factors associated with developing CP have been described as toxic
(e.g., alcohol and tobacco); idiopathic (e.g., unknown); genetic, autoimmune, recurrent
acute pancreatitis, and obstructive (the TIGAR-O system). Upon histological screening of
the pancreata from a cohort of CP patients who had undergone pancreatectomy for the
treatment of intractable pain in Leicester, UK, one sample showed a striking change in the
morphological balance toward an endocrine phenotype, most notably there was evidence of
substantial α cell genesis enveloping entire cross sections of ductal epithelium and the
presence of α cells within the ductal lumens. This patient had previously undergone a
partial pancreatectomy, had severe sclerosing CP, an exceptionally low body mass index
(15.2), and diabetes at the time the pancreas was removed, and although these factors have
been shown to induce tissue remodeling, such high levels of α cells was an unusual finding
within our series of patients. Due to the fact that α cells have been shown to be the
first endocrine cell type that emerges during islet neogenesis, future research profiling
the factors that caused such marked α cell genesis may prove useful in the field of islet
transplantation.
Collapse
Affiliation(s)
- M'Balu A Webb
- 1 The Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester General Hospital, Leicester, United Kingdom
| | - Jane J Chen
- 2 Department of Hepatobiliary Surgery, University Hospitals of Leicester NHS Trust, Leicester General Hospital, Leicester, United Kingdom
| | - Roger F L James
- 3 Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Melanie J Davies
- 1 The Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester General Hospital, Leicester, United Kingdom.,4 Diabetes Research Centre, University of Leicester, Leicester, United Kingdom
| | - Ashley R Dennison
- 2 Department of Hepatobiliary Surgery, University Hospitals of Leicester NHS Trust, Leicester General Hospital, Leicester, United Kingdom
| |
Collapse
|
73
|
Benomar K, Chetboun M, Espiard S, Jannin A, Le Mapihan K, Gmyr V, Caiazzo R, Torres F, Raverdy V, Bonner C, D'Herbomez M, Pigny P, Noel C, Kerr-Conte J, Pattou F, Vantyghem MC. Purity of islet preparations and 5-year metabolic outcome of allogenic islet transplantation. Am J Transplant 2018; 18:945-951. [PMID: 28941330 DOI: 10.1111/ajt.14514] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/23/2017] [Accepted: 09/04/2017] [Indexed: 01/25/2023]
Abstract
In allogenic islet transplantation (IT), high purity of islet preparations and low contamination by nonislet cells are generally favored. The aim of the present study was to analyze the relation between the purity of transplanted preparations and graft function during 5 years post-IT. Twenty-four patients with type 1 diabetes, followed for 5 years after IT, were enrolled. Metabolic parameters and daily insulin requirements were compared between patients who received islet preparations with a mean purity <50% (LOW purity) or ≥50% (HIGH purity). We also analyzed blood levels of carbohydrate antigen 19-9 (CA 19-9)-a biomarker of pancreatic ductal cells-and glucagon, before and after IT. At 5 years, mean hemoglobin A1c (HbA1c levels) (P = .01) and daily insulin requirements (P = .03) were lower in the LOW purity group. Insulin independence was more frequent in the LOW purity group (P < .05). CA19-9 and glucagon levels increased post-IT (P < .0001) and were inversely correlated with the degree of purity. Overall, our results suggest that nonislet cells have a beneficial effect on long-term islet graft function, possibly through ductal-to-endocrine cell differentiation. ClinicalTrial.gov NCT00446264 and NCT01123187.
Collapse
Affiliation(s)
- K Benomar
- Department of Endocrinology and Metabolism, CHRU Lille, Lille, France.,UMR 1190, Translational Research in Diabetes INSERM, Lille, France.,EGID (European Genomic Institute for Diabetes), Univ Lille, Lille, France
| | - M Chetboun
- UMR 1190, Translational Research in Diabetes INSERM, Lille, France.,EGID (European Genomic Institute for Diabetes), Univ Lille, Lille, France.,Department of Endocrine Surgery, CHRU Lille, Lille, France
| | - S Espiard
- Department of Endocrinology and Metabolism, CHRU Lille, Lille, France
| | - A Jannin
- Department of Endocrinology and Metabolism, CHRU Lille, Lille, France
| | - K Le Mapihan
- Department of Endocrinology and Metabolism, CHRU Lille, Lille, France
| | - V Gmyr
- UMR 1190, Translational Research in Diabetes INSERM, Lille, France.,EGID (European Genomic Institute for Diabetes), Univ Lille, Lille, France
| | - R Caiazzo
- UMR 1190, Translational Research in Diabetes INSERM, Lille, France.,EGID (European Genomic Institute for Diabetes), Univ Lille, Lille, France.,Department of Endocrine Surgery, CHRU Lille, Lille, France
| | - F Torres
- UMR 1190, Translational Research in Diabetes INSERM, Lille, France.,EGID (European Genomic Institute for Diabetes), Univ Lille, Lille, France.,Department of Endocrine Surgery, CHRU Lille, Lille, France
| | - V Raverdy
- UMR 1190, Translational Research in Diabetes INSERM, Lille, France.,EGID (European Genomic Institute for Diabetes), Univ Lille, Lille, France.,Department of Endocrine Surgery, CHRU Lille, Lille, France
| | - C Bonner
- UMR 1190, Translational Research in Diabetes INSERM, Lille, France.,EGID (European Genomic Institute for Diabetes), Univ Lille, Lille, France
| | - M D'Herbomez
- Department of Biology, CHRU Lille, Lille, France
| | - P Pigny
- Department of Biology, CHRU Lille, Lille, France
| | - C Noel
- Department of Nephrology and Transplantation, CHRU Lille, Lille, France
| | - J Kerr-Conte
- UMR 1190, Translational Research in Diabetes INSERM, Lille, France.,EGID (European Genomic Institute for Diabetes), Univ Lille, Lille, France
| | - F Pattou
- UMR 1190, Translational Research in Diabetes INSERM, Lille, France.,EGID (European Genomic Institute for Diabetes), Univ Lille, Lille, France.,Department of Endocrine Surgery, CHRU Lille, Lille, France
| | - M C Vantyghem
- Department of Endocrinology and Metabolism, CHRU Lille, Lille, France.,UMR 1190, Translational Research in Diabetes INSERM, Lille, France.,EGID (European Genomic Institute for Diabetes), Univ Lille, Lille, France
| |
Collapse
|
74
|
Kulkarni AA, Conteh AM, Sorrell CA, Mirmira A, Tersey SA, Mirmira RG, Linnemann AK, Anderson RM. An In Vivo Zebrafish Model for Interrogating ROS-Mediated Pancreatic β-Cell Injury, Response, and Prevention. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1324739. [PMID: 29785241 PMCID: PMC5896207 DOI: 10.1155/2018/1324739] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/23/2018] [Indexed: 01/10/2023]
Abstract
It is well known that a chronic state of elevated reactive oxygen species (ROS) in pancreatic β-cells impairs their ability to release insulin in response to elevated plasma glucose. Moreover, at its extreme, unmitigated ROS drives regulated cell death. This dysfunctional state of ROS buildup can result both from genetic predisposition and environmental factors such as obesity and overnutrition. Importantly, excessive ROS buildup may underlie metabolic pathologies such as type 2 diabetes mellitus. The ability to monitor ROS dynamics in β-cells in situ and to manipulate it via genetic, pharmacological, and environmental means would accelerate the development of novel therapeutics that could abate this pathology. Currently, there is a lack of models with these attributes that are available to the field. In this study, we use a zebrafish model to demonstrate that ROS can be generated in a β-cell-specific manner using a hybrid chemical genetic approach. Using a transgenic nitroreductase-expressing zebrafish line, Tg(ins:Flag-NTR)s950 , treated with the prodrug metronidazole (MTZ), we found that ROS is rapidly and explicitly generated in β-cells. Furthermore, the level of ROS generated was proportional to the dosage of prodrug added to the system. At high doses of MTZ, caspase 3 was rapidly cleaved, β-cells underwent regulated cell death, and macrophages were recruited to the islet to phagocytose the debris. Based on our findings, we propose a model for the mechanism of NTR/MTZ action in transgenic eukaryotic cells and demonstrate the robust utility of this system to model ROS-related disease pathology.
Collapse
Affiliation(s)
- Abhishek A. Kulkarni
- Center for Diabetes and Metabolic Disease, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Abass M. Conteh
- Center for Diabetes and Metabolic Disease, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Cody A. Sorrell
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Anjali Mirmira
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sarah A. Tersey
- Center for Diabetes and Metabolic Disease, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Raghavendra G. Mirmira
- Center for Diabetes and Metabolic Disease, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Amelia K. Linnemann
- Center for Diabetes and Metabolic Disease, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ryan M. Anderson
- Center for Diabetes and Metabolic Disease, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
75
|
da Silva ABS, Fonseca CMB, Cavalcante MMADS, de Oliveira IM, Ferraz MS, Viana FJC, Fontenele RD, Conde Júnior AM. Histomorphometry of pancreas development in hybrid chicken (Galus galus) embryo and fetus. Microsc Res Tech 2018. [PMID: 29527773 DOI: 10.1002/jemt.23016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The pancreas comprises an important metabolic organ of endocrine and exocrine character that has embryonic origin of rudimentary buds that fuse to form the organ. The present work aims to describe the pancreatic histogenesis of hybrid chick embryos (Gallus gallus). The research was performed in the UFPI, previously approved by the CEUA with protocol no. 040/15. We used 120 fertilized eggs of hybrid chickens kept in an incubator with controlled temperature and humidity. Daily collections of embryos and fetuses were performed from 4 to 21 days of incubation through the anatomical dissection consecutive the euthanasia. The tissues, previously fixed in 10% buffered formaldehyde, were submitted to histological processing and stained with hematoxylin-eosin. Finally, the mounted slides were analyzed in image software to obtain histomorphometric data, which were submitted to statistical analysis. The pancreas of hybrid chicken embryos originates around the fourth day of incubation with the dorsal and ventral pancreatic bud formation, which are composed by epithelial and mesenchymal cells. These cells differ in exocrine and endocrine cells. Around twelve embryonic days occurs the buds fusion and the immature organ formation that will give continue with the ductal system development, vascularization and compartmentalization of the endocrine and exocrine parts. Until 21st day of incubation it is possible to identify undifferentiated tissue forms which suggesting postnatal histogenesis. The description of pancreas histogenesis using histometric data on hybrid chicken embryos contributes to the clarification of embryonic development and reaffirms the premise that chickens serve as an experimental model for embryonic study of mammals.
Collapse
|
76
|
Saitoh S, Ohno N, Saitoh Y, Terada N, Shimo S, Aida K, Fujii H, Kobayashi T, Ohno S. Improved Serial Sectioning Techniques for Correlative Light-Electron Microscopy Mapping of Human Langerhans Islets. Acta Histochem Cytochem 2018; 51:9-20. [PMID: 29622846 PMCID: PMC5880804 DOI: 10.1267/ahc.17020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 11/29/2017] [Indexed: 01/19/2023] Open
Abstract
Combined analysis of immunostaining for various biological molecules coupled with investigations of ultrastructural features of individual cells is a powerful approach for studies of cellular functions in normal and pathological conditions. However, weak antigenicity of tissues fixed by conventional methods poses a problem for immunoassays. This study introduces a method of correlative light and electron microscopy imaging of the same endocrine cells of compact and diffuse islets from human pancreatic tissue specimens. The method utilizes serial sections obtained from Epon-embedded specimens fixed with glutaraldehyde and osmium tetroxide. Double-immunofluorescence staining of thick Epon sections for endocrine hormones (insulin and glucagon) and regenerating islet-derived gene 1 α (REG1α) was performed following the removal of Epoxy resin with sodium ethoxide, antigen retrieval by autoclaving, and de-osmification treatment with hydrogen peroxide. The immunofluorescence images of endocrine cells were superimposed with the electron microscopy images of the same cells obtained from serial ultrathin sections. Immunofluorescence images showed well-preserved secretory granules in endocrine cells, whereas electron microscopy observations demonstrated corresponding secretory granules and intracellular organelles in the same cells. In conclusion, the correlative imaging approach developed by us may be useful for examining ultrastructural features in combination with immunolocalisation of endocrine hormones in the same human pancreatic islets.
Collapse
Affiliation(s)
- Sei Saitoh
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi
- Present address: Section of Electron Microscopy, Supportive Center for Brain Research, National Institute for Physiological Sciences
| | - Nobuhiko Ohno
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi
| | - Yurika Saitoh
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi
| | - Nobuo Terada
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi
- Department of Occupational Therapy, School of Health Sciences, Shinshu University School of Medicine
| | - Satoshi Shimo
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi
| | - Kaoru Aida
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi
| | - Hideki Fujii
- First Department of Surgery, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi
| | - Tetsuro Kobayashi
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi
| | - Shinichi Ohno
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi
| |
Collapse
|
77
|
Wyett G, Gibert Y, Ellis M, Castillo HA, Aston-Mourney K. Metformin, beta-cell development, and novel processes following beta-cell ablation in zebrafish. Endocrine 2018; 59:419-425. [PMID: 29274062 DOI: 10.1007/s12020-017-1502-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 12/12/2017] [Indexed: 11/25/2022]
Abstract
PURPOSE Type 1 and 2 diabetes are characterized by a loss of insulin-producing beta-cells. Current treatments help maintain blood glucose levels but cannot provide a cure. As such, a vital target for the cure of diabetes is a way to restore beta-cell mass. The drug metformin can protect cultured beta-cells/islets from hyperglycemia-induced dysfunction and death. Further, treatment of pregnant mice with metformin results in an enhanced beta-cell fraction in the embryos; however, whether this occurs via a direct effect is unknown. METHODS We utilized the external embryogenesis of the zebrafish to determine the direct effect of metformin treatment on the pancreas of the developing embryo and following beta-cell ablation. RESULTS During development metformin did not alter beta-cell or alpha-cell mass but had a small effect to increase delta-cell mass as measured by in situ hybridization. Further metformin significantly increased beta-cell number. Following beta-cell ablation, both glucagon and somatostatin expression were upregulated (>2-fold). Additionally, while metformin showed no effect to alter beta-cell mass or number, somatostatin expression was further increased (>5-fold). CONCLUSIONS We showed that direct exposure to metformin during embryogenesis does not increase insulin-expressing area but does increase beta-cell number. Further, we identified novel consequences of beta-cell ablation to alter the expression of other pancreatic hormones that were enhanced by metformin. Therefore, this study provides a greater understanding of the beta-cell development/regenerative processes and the effect of metformin, bringing us closer to identifying how to increase beta-cells in humans.
Collapse
Affiliation(s)
- Georgia Wyett
- Metabolic Research Unit, School of Medicine, Deakin University, 75 Pigdons Rd, Waurn Ponds, VIC, 3216, Australia
| | - Yann Gibert
- Metabolic Research Unit, School of Medicine, Deakin University, 75 Pigdons Rd, Waurn Ponds, VIC, 3216, Australia
| | - Megan Ellis
- Metabolic Research Unit, School of Medicine, Deakin University, 75 Pigdons Rd, Waurn Ponds, VIC, 3216, Australia
| | - Hozana A Castillo
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
- Brazilian Biosciences National Laboratory, Brazilian Center of Research in Energy and Materials, Campinas, Brazil
| | - Kathryn Aston-Mourney
- Metabolic Research Unit, School of Medicine, Deakin University, 75 Pigdons Rd, Waurn Ponds, VIC, 3216, Australia.
| |
Collapse
|
78
|
Abstract
Type 1 diabetes is characterized by selective loss of beta cells and insulin secretion, which significantly impact glucose homeostasis. However, this progressive disease is also associated with dysfunction of the alpha cell component of the islet, which can exacerbate hyperglycemia due to paradoxical hyperglucagonemia or lead to severe hypoglycemia as a result of failed counterregulation. In this review, the physiology of alpha cell secretion and the potential mechanisms underlying alpha cell dysfunction in type 1 diabetes will be explored. Because type 1 diabetes is a progressive disease, a synthesized timeline of aberrant alpha cell function will be presented as an attempt to delineate the natural history of type 1 diabetes with respect to the alpha cell.
Collapse
Affiliation(s)
- Gina L C Yosten
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Saint Louis, MO 63104, United States.
| |
Collapse
|
79
|
Kamel M, Ninov N. Catching new targets in metabolic disease with a zebrafish. Curr Opin Pharmacol 2017; 37:41-50. [DOI: 10.1016/j.coph.2017.08.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 08/04/2017] [Accepted: 08/11/2017] [Indexed: 12/12/2022]
|
80
|
Different developmental histories of beta-cells generate functional and proliferative heterogeneity during islet growth. Nat Commun 2017; 8:664. [PMID: 28939870 PMCID: PMC5610262 DOI: 10.1038/s41467-017-00461-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/30/2017] [Indexed: 12/11/2022] Open
Abstract
The proliferative and functional heterogeneity among seemingly uniform cells is a universal phenomenon. Identifying the underlying factors requires single-cell analysis of function and proliferation. Here we show that the pancreatic beta-cells in zebrafish exhibit different growth-promoting and functional properties, which in part reflect differences in the time elapsed since birth of the cells. Calcium imaging shows that the beta-cells in the embryonic islet become functional during early zebrafish development. At later stages, younger beta-cells join the islet following differentiation from post-embryonic progenitors. Notably, the older and younger beta-cells occupy different regions within the islet, which generates topological asymmetries in glucose responsiveness and proliferation. Specifically, the older beta-cells exhibit robust glucose responsiveness, whereas younger beta-cells are more proliferative but less functional. As the islet approaches its mature state, heterogeneity diminishes and beta-cells synchronize function and proliferation. Our work illustrates a dynamic model of heterogeneity based on evolving proliferative and functional beta-cell states. Βeta-cells have recently been shown to be heterogeneous with regard to morphology and function. Here, the authors show that β-cells in zebrafish switch from proliferative to functional states with increasing time since β-cell birth, leading to functional and proliferative heterogeneity.
Collapse
|
81
|
Mondal P, Prasad A, Girdhar K. Interventions to improve β-cell mass and function. ANNALES D'ENDOCRINOLOGIE 2017; 78:469-477. [PMID: 28870707 DOI: 10.1016/j.ando.2016.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/18/2016] [Accepted: 11/09/2016] [Indexed: 01/09/2023]
Abstract
Diabetes mellitus (T2DM) has become an epidemiologically important disease worldwide and is also becoming a great matter of concern due to the effects associated with it like: high morbidity, elevated health care cost and shortened life span. T2DM is a chronic metabolic disease characterized by insulin resistance as well as β-cell dysfunction. It is widely accepted that in the face of insulin resistance, euglycemia can be maintained by increase in pancreatic β-cell mass and insulin secretion. This compensation is largely due to enhanced secretion of insulin by the β-cell mass, which is present initially, and thereby subsequent increases in β-cell mass provide additional insulin secretion. However, the mechanism by which β-cell anatomical plasticity and functional plasticity for insulin secretion is coordinated and executed in different physiological and pathophysiological states is complex and has been poorly understood. As the incidence of T2DM continues to increase at an alarming rate, it is becoming imperative to shift the research focus towards the β-cell physiology where identification of novel pathways that influence the β-cell proliferation and/or contribute to increase insulin secretion has the potential to lead to new therapies for preventing or delaying onset of disease.
Collapse
Affiliation(s)
- Prosenjit Mondal
- School of Basic Sciences, BioX, Indian Institute of Technology, Mandi, HP 175005, India.
| | - Amit Prasad
- School of Basic Sciences, BioX, Indian Institute of Technology, Mandi, HP 175005, India
| | - Khyati Girdhar
- School of Basic Sciences, BioX, Indian Institute of Technology, Mandi, HP 175005, India
| |
Collapse
|
82
|
Tritschler S, Theis FJ, Lickert H, Böttcher A. Systematic single-cell analysis provides new insights into heterogeneity and plasticity of the pancreas. Mol Metab 2017; 6:974-990. [PMID: 28951822 PMCID: PMC5605721 DOI: 10.1016/j.molmet.2017.06.021] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/13/2017] [Accepted: 06/19/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Diabetes mellitus is characterized by loss or dysfunction of insulin-producing β-cells in the pancreas, resulting in failure of blood glucose regulation and devastating secondary complications. Thus, β-cells are currently the prime target for cell-replacement and regenerative therapy. Triggering endogenous repair is a promising strategy to restore β-cell mass and normoglycemia in diabetic patients. Potential strategies include targeting specific β-cell subpopulations to increase proliferation or maturation. Alternatively, transdifferentiation of pancreatic islet cells (e.g. α- or δ-cells), extra-islet cells (acinar and ductal cells), hepatocytes, or intestinal cells into insulin-producing cells might improve glycemic control. To this end, it is crucial to systematically characterize and unravel the transcriptional program of all pancreatic cell types at the molecular level in homeostasis and disease. Furthermore, it is necessary to better determine the underlying mechanisms of β-cell maturation, maintenance, and dysfunction in diabetes, to identify and molecularly profile endocrine subpopulations with regenerative potential, and to translate the findings from mice to man. Recent approaches in single-cell biology started to illuminate heterogeneity and plasticity in the pancreas that might be targeted for β-cell regeneration in diabetic patients. SCOPE OF REVIEW This review discusses recent literature on single-cell analysis including single-cell RNA sequencing, single-cell mass cytometry, and flow cytometry of pancreatic cell types in the context of mechanisms of endogenous β-cell regeneration. We discuss new findings on the regulation of postnatal β-cell proliferation and maturation. We highlight how single-cell analysis recapitulates described principles of functional β-cell heterogeneity in animal models and adds new knowledge on the extent of β-cell heterogeneity in humans as well as its role in homeostasis and disease. Furthermore, we summarize the findings on cell subpopulations with regenerative potential that might enable the formation of new β-cells in diseased state. Finally, we review new data on the transcriptional program and function of rare pancreatic cell types and their implication in diabetes. MAJOR CONCLUSION Novel, single-cell technologies offer high molecular resolution of cellular heterogeneity within the pancreas and provide information on processes and factors that govern β-cell homeostasis, proliferation, and maturation. Eventually, these technologies might lead to the characterization of cells with regenerative potential and unravel disease-associated changes in gene expression to identify cellular and molecular targets for therapy.
Collapse
Affiliation(s)
- Sophie Tritschler
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Am Parkring 11, 85748 Garching-Hochbrück, Germany
- German Center for Diabetes Research, 85764 Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Fabian J. Theis
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Am Parkring 11, 85748 Garching-Hochbrück, Germany
- German Center for Diabetes Research, 85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Anika Böttcher
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Am Parkring 11, 85748 Garching-Hochbrück, Germany
- German Center for Diabetes Research, 85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| |
Collapse
|
83
|
Tarifeño-Saldivia E, Lavergne A, Bernard A, Padamata K, Bergemann D, Voz ML, Manfroid I, Peers B. Transcriptome analysis of pancreatic cells across distant species highlights novel important regulator genes. BMC Biol 2017; 15:21. [PMID: 28327131 PMCID: PMC5360028 DOI: 10.1186/s12915-017-0362-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/01/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Defining the transcriptome and the genetic pathways of pancreatic cells is of great interest for elucidating the molecular attributes of pancreas disorders such as diabetes and cancer. As the function of the different pancreatic cell types has been maintained during vertebrate evolution, the comparison of their transcriptomes across distant vertebrate species is a means to pinpoint genes under strong evolutionary constraints due to their crucial function, which have therefore preserved their selective expression in these pancreatic cell types. RESULTS In this study, RNA-sequencing was performed on pancreatic alpha, beta, and delta endocrine cells as well as the acinar and ductal exocrine cells isolated from adult zebrafish transgenic lines. Comparison of these transcriptomes identified many novel markers, including transcription factors and signaling pathway components, specific for each cell type. By performing interspecies comparisons, we identified hundreds of genes with conserved enriched expression in endocrine and exocrine cells among human, mouse, and zebrafish. This list includes many genes known as crucial for pancreatic cell formation or function, but also pinpoints many factors whose pancreatic function is still unknown. A large set of endocrine-enriched genes can already be detected at early developmental stages as revealed by the transcriptomic profiling of embryonic endocrine cells, indicating a potential role in cell differentiation. The actual involvement of conserved endocrine genes in pancreatic cell differentiation was demonstrated in zebrafish for myt1b, whose invalidation leads to a reduction of alpha cells, and for cdx4, selectively expressed in endocrine delta cells and crucial for their specification. Intriguingly, comparison of the endocrine alpha and beta cell subtypes from human, mouse, and zebrafish reveals a much lower conservation of the transcriptomic signatures for these two endocrine cell subtypes compared to the signatures of pan-endocrine and exocrine cells. These data suggest that the identity of the alpha and beta cells relies on a few key factors, corroborating numerous examples of inter-conversion between these two endocrine cell subtypes. CONCLUSION This study highlights both evolutionary conserved and species-specific features that will help to unveil universal and fundamental regulatory pathways as well as pathways specific to human and laboratory animal models such as mouse and zebrafish.
Collapse
Affiliation(s)
- Estefania Tarifeño-Saldivia
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium
| | - Arnaud Lavergne
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium
| | - Alice Bernard
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium
| | - Keerthana Padamata
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium
| | - David Bergemann
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium
| | - Marianne L Voz
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium
| | - Isabelle Manfroid
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium
| | - Bernard Peers
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l'Hôpital 1, B34, 4000 Sart Tilman, Liege, Belgium.
| |
Collapse
|
84
|
Maddison LA, Chen W. Modeling Pancreatic Endocrine Cell Adaptation and Diabetes in the Zebrafish. Front Endocrinol (Lausanne) 2017; 8:9. [PMID: 28184214 PMCID: PMC5266698 DOI: 10.3389/fendo.2017.00009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 01/11/2017] [Indexed: 12/13/2022] Open
Abstract
Glucose homeostasis is an important element of energy balance and is conserved in organisms from fruit fly to mammals. Central to the control of circulating glucose levels in vertebrates are the endocrine cells of the pancreas, particularly the insulin-producing β-cells and the glucagon producing α-cells. A feature of α- and β-cells is their plasticity, an ability to adapt, in function and number as a response to physiological and pathophysiological conditions of increased hormone demand. The molecular mechanisms underlying these adaptive responses that maintain glucose homeostasis are incompletely defined. The zebrafish is an attractive model due to the low cost, high fecundity, and amenability to genetic and compound screens, and mechanisms governing the development of the pancreatic endocrine cells are conserved between zebrafish and mammals. Post development, both β- and α-cells of zebrafish display plasticity as in mammals. Here, we summarize the studies of pancreatic endocrine cell adaptation in zebrafish. We further explore the utility of the zebrafish as a model for diabetes, a relevant topic considering the increase in diabetes in the human population.
Collapse
Affiliation(s)
- Lisette A. Maddison
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Wenbiao Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
85
|
Abstract
The zebrafish pancreas shares its basic organization and cell types with the mammalian pancreas. In addition, the developmental pathways that lead to the establishment of the pancreatic islets of Langherhans are generally conserved from fish to mammals. Zebrafish provides a powerful tool to probe the mechanisms controlling establishment of the pancreatic endocrine cell types from early embryonic progenitor cells, as well as the regeneration of endocrine cells after damage. This knowledge is, in turn, applicable to refining protocols to generate renewable sources of human pancreatic islet cells that are critical for regulation of blood sugar levels. Here, we review how previous and ongoing studies in zebrafish and beyond are influencing the understanding of molecular mechanisms underlying various forms of diabetes and efforts to develop cell-based approaches to cure this increasingly widespread disease.
Collapse
|
86
|
Li J, Casteels T, Frogne T, Ingvorsen C, Honoré C, Courtney M, Huber KVM, Schmitner N, Kimmel RA, Romanov RA, Sturtzel C, Lardeau CH, Klughammer J, Farlik M, Sdelci S, Vieira A, Avolio F, Briand F, Baburin I, Májek P, Pauler FM, Penz T, Stukalov A, Gridling M, Parapatics K, Barbieux C, Berishvili E, Spittler A, Colinge J, Bennett KL, Hering S, Sulpice T, Bock C, Distel M, Harkany T, Meyer D, Superti-Furga G, Collombat P, Hecksher-Sørensen J, Kubicek S. Artemisinins Target GABA A Receptor Signaling and Impair α Cell Identity. Cell 2016; 168:86-100.e15. [PMID: 27916275 PMCID: PMC5236063 DOI: 10.1016/j.cell.2016.11.010] [Citation(s) in RCA: 299] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 08/04/2016] [Accepted: 11/03/2016] [Indexed: 12/12/2022]
Abstract
Type 1 diabetes is characterized by the destruction of pancreatic β cells, and generating new insulin-producing cells from other cell types is a major aim of regenerative medicine. One promising approach is transdifferentiation of developmentally related pancreatic cell types, including glucagon-producing α cells. In a genetic model, loss of the master regulatory transcription factor Arx is sufficient to induce the conversion of α cells to functional β-like cells. Here, we identify artemisinins as small molecules that functionally repress Arx by causing its translocation to the cytoplasm. We show that the protein gephyrin is the mammalian target of these antimalarial drugs and that the mechanism of action of these molecules depends on the enhancement of GABAA receptor signaling. Our results in zebrafish, rodents, and primary human pancreatic islets identify gephyrin as a druggable target for the regeneration of pancreatic β cell mass from α cells. Artemisinins inhibit ARX function and impair α cell identity Compounds act by stabilizing gephyrin, thus enhancing GABAA receptor signaling Artemisinins increase β cell mass in zebrafish and rodent models Functional and transcriptional data indicate a conserved phenotype in human islets
Collapse
Affiliation(s)
- Jin Li
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria
| | - Tamara Casteels
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria
| | - Thomas Frogne
- Novo Nordisk A/S, Novo Nordisk Park, DK-2760 Måløv, Denmark
| | | | | | - Monica Courtney
- Université Côte d'Azur, INSERM, CNRS, iBV, 06108 Nice, France
| | - Kilian V M Huber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria
| | - Nicole Schmitner
- Institute of Molecular Biology, Leopold-Franzens-University Innsbruck, Technikerstr. 25, 6020 Innsbruck, Austria
| | - Robin A Kimmel
- Institute of Molecular Biology, Leopold-Franzens-University Innsbruck, Technikerstr. 25, 6020 Innsbruck, Austria
| | - Roman A Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria; Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Caterina Sturtzel
- Children's Cancer Research Institute, Innovative Cancer Models, Zimmermannplatz 10, 1090 Vienna, Austria
| | - Charles-Hugues Lardeau
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria; Christian Doppler Laboratory for Chemical Epigenetics and Antiinfectives, CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, 1090 Vienna, Austria
| | - Johanna Klughammer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria
| | - Matthias Farlik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria
| | - Sara Sdelci
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria
| | - Andhira Vieira
- Université Côte d'Azur, INSERM, CNRS, iBV, 06108 Nice, France
| | - Fabio Avolio
- Université Côte d'Azur, INSERM, CNRS, iBV, 06108 Nice, France
| | - François Briand
- Physiogenex S.A.S., Prologue Biotech, 516, rue Pierre et Marie Curie, 31670 Labege, France
| | - Igor Baburin
- Institute of Pharmacology and Toxicology, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Peter Májek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria
| | - Florian M Pauler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria
| | - Thomas Penz
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria
| | - Alexey Stukalov
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria
| | - Manuela Gridling
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria
| | - Katja Parapatics
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria
| | - Charlotte Barbieux
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, 1211 Geneva, Switzerland
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, 1211 Geneva, Switzerland; Institute of Medical Research, Ilia State University, Tbilisi 0162, Georgia
| | - Andreas Spittler
- Core Facility Flow Cytometry and Department of Surgery, Research Laboratories, Medical University of Vienna, 1090 Vienna, Austria
| | - Jacques Colinge
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria
| | - Keiryn L Bennett
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria
| | - Steffen Hering
- Institute of Pharmacology and Toxicology, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Thierry Sulpice
- Physiogenex S.A.S., Prologue Biotech, 516, rue Pierre et Marie Curie, 31670 Labege, France
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria; Max Planck Institute for Informatics, 66123 Saarbrücken, Germany
| | - Martin Distel
- Children's Cancer Research Institute, Innovative Cancer Models, Zimmermannplatz 10, 1090 Vienna, Austria
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria; Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Dirk Meyer
- Institute of Molecular Biology, Leopold-Franzens-University Innsbruck, Technikerstr. 25, 6020 Innsbruck, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria; Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | | | | | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences. Lazarettgasse 14, 1090 Vienna, Austria; Christian Doppler Laboratory for Chemical Epigenetics and Antiinfectives, CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, 1090 Vienna, Austria.
| |
Collapse
|
87
|
Wang YJ, Golson ML, Schug J, Traum D, Liu C, Vivek K, Dorrell C, Naji A, Powers AC, Chang KM, Grompe M, Kaestner KH. Single-Cell Mass Cytometry Analysis of the Human Endocrine Pancreas. Cell Metab 2016; 24:616-626. [PMID: 27732837 PMCID: PMC5123805 DOI: 10.1016/j.cmet.2016.09.007] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/26/2016] [Accepted: 09/21/2016] [Indexed: 12/30/2022]
Abstract
The human endocrine pancreas consists of multiple cell types and plays a critical role in glucose homeostasis. Here, we apply mass cytometry technology to measure all major islet hormones, proliferative markers, and readouts of signaling pathways involved in proliferation at single-cell resolution. Using this innovative technology, we simultaneously examined baseline proliferation levels of all endocrine cell types from birth through adulthood, as well as in response to the mitogen harmine. High-dimensional analysis of our marker protein expression revealed three major clusters of beta cells within individuals. Proliferating beta cells are confined to two of the clusters.
Collapse
Affiliation(s)
- Yue J Wang
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria L Golson
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan Schug
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel Traum
- Medical Research, Corporal Michael J. Crescenz Veterans Affairs Medical Center and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chengyang Liu
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kumar Vivek
- Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10467, USA
| | - Craig Dorrell
- Oregon Stem Cell Center, Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Ali Naji
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alvin C Powers
- Departments of Molecular Physiology and Biophysics and Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 19147, USA; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Kyong-Mi Chang
- Medical Research, Corporal Michael J. Crescenz Veterans Affairs Medical Center and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Markus Grompe
- Oregon Stem Cell Center, Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
88
|
O'Harte FPM, Ng MT, Lynch AM, Conlon JM, Flatt PR. Dogfish glucagon analogues counter hyperglycaemia and enhance both insulin secretion and action in diet-induced obese diabetic mice. Diabetes Obes Metab 2016; 18:1013-24. [PMID: 27357054 DOI: 10.1111/dom.12713] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/16/2016] [Accepted: 06/23/2016] [Indexed: 12/11/2022]
Abstract
AIMS To investigate the antidiabetic actions of three dogfish glucagon peptide analogues [known glucagon-like peptide-1 and glucagon receptor co-agonists] after chronic administration in diet-induced high-fat-diet-fed diabetic mice. MATERIALS AND METHODS National Institutes of Health Swiss mice were pre-conditioned to a high-fat diet (45% fat) for 100 days, and control mice were fed a normal diet (10% fat). Normal diet control and high-fat-fed control mice received twice-daily intraperitoneal (i.p.) saline injections, while the high-fat-fed treatment groups (n = 8) received twice-daily injections of exendin-4(1-39), [S2a]dogfish glucagon, [S2a]dogfish glucagon exendin-4(31-39) or [S2a]dogfish glucagon-Lys(30) -γ-glutamyl-PAL (25 nmol/kg body weight) for 51 days. RESULTS After dogfish glucagon analogue treatment, there was a rapid and sustained decrease in non-fasting blood glucose and an associated insulinotropic effect (analysis of variance, p < .05 to <.001) compared with saline-treated high-fat-fed controls. All peptide treatments significantly improved i.p. and oral glucose tolerance with concomitant increased insulin secretion compared with saline-treated high-fat-fed controls (p <.05 to <.001). After chronic treatment, no receptor desensitization was observed but insulin sensitivity was enhanced for all peptide-treated groups (p < .01 to <.001) except [S2a]dogfish glucagon. Both exendin-4 and [S2a]dogfish glucagon exendin-4(31-39) significantly reduced plasma triglyceride concentrations compared with those found in lean controls (p = .0105 and p = .0048, respectively). Pancreatic insulin content was not affected by peptide treatments but [S2a]dogfish glucagon and [S2a]dogfish glucagon exendin-4(31-39) decreased pancreatic glucagon by 28%-34% (p = .0221 and p = .0075, respectively). The percentage of β-cell area within islets was increased by exendin-4 and peptide analogue treatment groups compared with high-fat-fed controls and the β-cell area decreased (p < .05 to <.01). CONCLUSIONS Overall, dogfish glucagon co-agonist analogues had several beneficial metabolic effects, showing therapeutic potential for type 2 diabetes.
Collapse
Affiliation(s)
- F P M O'Harte
- School of Biomedical Sciences, Saad Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, UK.
| | - M T Ng
- School of Biomedical Sciences, Saad Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, UK
| | - A M Lynch
- School of Biomedical Sciences, Saad Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, UK
| | - J M Conlon
- School of Biomedical Sciences, Saad Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, UK
| | - P R Flatt
- School of Biomedical Sciences, Saad Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, UK
| |
Collapse
|
89
|
Abu-Farha M, Al Madhoun A, Abubaker J. The Rise and the Fall of Betatrophin/ANGPTL8 as an Inducer of β-Cell Proliferation. J Diabetes Res 2016; 2016:4860595. [PMID: 27672665 PMCID: PMC5031879 DOI: 10.1155/2016/4860595] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 12/14/2022] Open
Abstract
Diabetes is a global health problem that is caused by impaired insulin production from pancreatic β-cells. Efforts to regenerate β-cells have been advancing rapidly in the past two decades with progress made towards identifying new agents that induce β-cells regeneration. ANGPTL8, also named betatrophin, has been recently identified as a hormone capable of inducing β-cells proliferation and increasing β-cells mass in rodents. Its discovery has been cherished as a breakthrough and a game changer in the field of β-cells regeneration. Initially, ANGPTL8 has been identified as atypical member of the angiopoietin-like protein family as a regulator of triglyceride in plasma through its interaction with ANGPTL3 and its regulation of lipoprotein lipase activity. In this review, we will review literature on the proposed role of ANGPTL8 in β-cells proliferation, the controversy regarding this role, and the emerging data questioning its involvement in β-cells proliferation. Additionally we will discuss new clinical data that describes its role in diabetes and the putative therapeutic targeting of this protein.
Collapse
Affiliation(s)
- Mohamed Abu-Farha
- Biochemistry and Molecular Biology Unit, Dasman Diabetes Institute, Kuwait City, Kuwait
| | | | - Jehad Abubaker
- Biochemistry and Molecular Biology Unit, Dasman Diabetes Institute, Kuwait City, Kuwait
| |
Collapse
|
90
|
Cigliola V, Thorel F, Chera S, Herrera PL. Stress-induced adaptive islet cell identity changes. Diabetes Obes Metab 2016; 18 Suppl 1:87-96. [PMID: 27615136 PMCID: PMC5021189 DOI: 10.1111/dom.12726] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 04/22/2016] [Indexed: 12/12/2022]
Abstract
The different forms of diabetes mellitus differ in their pathogenesis but, ultimately, they are all characterized by progressive islet β-cell loss. Restoring the β-cell mass is therefore a major goal for future therapeutic approaches. The number of β-cells found at birth is determined by proliferation and differentiation of pancreatic progenitor cells, and it has been considered to remain mostly unchanged throughout adult life. Recent studies in mice have revealed an unexpected plasticity in islet endocrine cells in response to stress; under certain conditions, islet non-β-cells have the potential to reprogram into insulin producers, thus contributing to restore the β-cell mass. Here, we discuss the latest findings on pancreas and islet cell plasticity upon physiological, pathological and experimental conditions of stress. Understanding the mechanisms involved in cell reprogramming in these models will allow the development of new strategies for the treatment of diabetes, by exploiting the intrinsic regeneration capacity of the pancreas.
Collapse
Affiliation(s)
- V Cigliola
- Department of Genetic Medicine and Development, Faculty of Medicine, Institute of Genetics and Genomics in Geneva (iGE3), and Centre facultaire du diabète, University of Geneva, Geneva, Switzerland
| | - F Thorel
- Department of Genetic Medicine and Development, Faculty of Medicine, Institute of Genetics and Genomics in Geneva (iGE3), and Centre facultaire du diabète, University of Geneva, Geneva, Switzerland
| | - S Chera
- Department of Clinical Science, Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
| | - P L Herrera
- Department of Genetic Medicine and Development, Faculty of Medicine, Institute of Genetics and Genomics in Geneva (iGE3), and Centre facultaire du diabète, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
91
|
O'Harte FPM, Ng MT, Lynch AM, Conlon JM, Flatt PR. Novel dual agonist peptide analogues derived from dogfish glucagon show promising in vitro insulin releasing actions and antihyperglycaemic activity in mice. Mol Cell Endocrinol 2016; 431:133-44. [PMID: 27179756 DOI: 10.1016/j.mce.2016.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/10/2016] [Accepted: 05/10/2016] [Indexed: 02/07/2023]
Abstract
The antidiabetic potential of thirteen novel dogfish glucagon derived analogues were assessed in vitro and in acute in vivo studies. Stable peptide analogues enhanced insulin secretion from BRIN-BD11 β-cells (p < 0.001) and reduced acute glycaemic responses following intraperitoneal glucose (25 nmol/kg) in healthy NIH Swiss mice (p < 0.05-p<0.001). The in vitro insulinotropic actions of [S2a]dogfish glucagon, [S2a]dogfish glucagon-exendin-4(31-39) and [S2a]dogfish glucagon-Lys(30)-γ-glutamyl-PAL, were blocked (p < 0.05-p<0.001) by the specific GLP-1 and glucagon receptor antagonists, exendin-4(9-39) and (desHis(1)Pro(4)Glu(9))glucagon amide but not by (Pro(3))GIP, indicating lack of GIP receptor involvement. These analogues dose-dependently stimulated cAMP production in GLP-1 and glucagon (p < 0.05-p<0.001) but not GIP-receptor transfected cells. They improved acute glycaemic and insulinotropic responses in high-fat fed diabetic mice and in wild-type C57BL/6J and GIPR-KO mice (p < 0.05-p<0.001), but not GLP-1R-KO mice, confirming action on GLP-1 but not GIP receptors. Overall, dogfish glucagon analogues have potential for diabetes therapy, exerting beneficial metabolic effects via GLP-1 and glucagon receptors.
Collapse
Affiliation(s)
- F P M O'Harte
- The Saad Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, Co. Derry, BT52 1SA, Northern Ireland, UK.
| | - M T Ng
- The Saad Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, Co. Derry, BT52 1SA, Northern Ireland, UK
| | - A M Lynch
- The Saad Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, Co. Derry, BT52 1SA, Northern Ireland, UK
| | - J M Conlon
- The Saad Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, Co. Derry, BT52 1SA, Northern Ireland, UK
| | - P R Flatt
- The Saad Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, Co. Derry, BT52 1SA, Northern Ireland, UK
| |
Collapse
|
92
|
Lu J, Liu KC, Schulz N, Karampelias C, Charbord J, Hilding A, Rautio L, Bertolino P, Östenson CG, Brismar K, Andersson O. IGFBP1 increases β-cell regeneration by promoting α- to β-cell transdifferentiation. EMBO J 2016; 35:2026-44. [PMID: 27516442 PMCID: PMC5116948 DOI: 10.15252/embj.201592903] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 06/27/2016] [Indexed: 01/09/2023] Open
Abstract
There is great interest in therapeutically harnessing endogenous regenerative mechanisms to increase the number of β cells in people with diabetes. By performing whole‐genome expression profiling of zebrafish islets, we identified 11 secreted proteins that are upregulated during β‐cell regeneration. We then tested the proteins' ability to potentiate β‐cell regeneration in zebrafish at supraphysiological levels. One protein, insulin‐like growth factor (Igf) binding‐protein 1 (Igfbp1), potently promoted β‐cell regeneration by potentiating α‐ to β‐cell transdifferentiation. Using various inhibitors and activators of the Igf pathway, we show that Igfbp1 exerts its regenerative effect, at least partly, by inhibiting Igf signaling. Igfbp1's effect on transdifferentiation appears conserved across species: Treating mouse and human islets with recombinant IGFBP1 in vitro increased the number of cells co‐expressing insulin and glucagon threefold. Moreover, a prospective human study showed that having high IGFBP1 levels reduces the risk of developing type‐2 diabetes by more than 85%. Thus, we identify IGFBP1 as an endogenous promoter of β‐cell regeneration and highlight its clinical importance in diabetes.
Collapse
Affiliation(s)
- Jing Lu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ka-Cheuk Liu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Nadja Schulz
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Christos Karampelias
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jérémie Charbord
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Hilding
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden
| | - Linn Rautio
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Philippe Bertolino
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR5286, Université Lyon 1, Lyon, France
| | - Claes-Göran Östenson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden
| | - Kerstin Brismar
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
93
|
Dash SN, Hakonen E, Ustinov J, Otonkoski T, Andersson O, Lehtonen S. sept7b is required for the differentiation of pancreatic endocrine progenitors. Sci Rep 2016; 6:24992. [PMID: 27114183 PMCID: PMC4845001 DOI: 10.1038/srep24992] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 04/05/2016] [Indexed: 12/14/2022] Open
Abstract
Protection or restoration of pancreatic β-cell mass as a therapeutic treatment for type 1 diabetes requires understanding of the mechanisms that drive the specification and development of pancreatic endocrine cells. Septins are filamentous small GTPases that function in the regulation of cell division, cytoskeletal organization and membrane remodeling, and are involved in various tissue-specific developmental processes. However, their role in pancreatic endocrine cell differentiation remains unknown. Here we show by functional manipulation techniques in transgenic zebrafish lines that suppression of sept7b, the zebrafish ortholog of human SEPT7, profoundly increases the number of endocrine progenitors but limits their differentiation, leading to reduction in β- and α-cell mass. Furthermore, we discovered that shh (sonic hedgehog) expression in the endoderm, essential for the development of pancreatic progenitors of the dorsal pancreatic bud, is absent in larvae depleted of sept7b. We also discovered that sept7b is important for the differentiation of ventral pancreatic bud-derived cells: sept7b-depleted larvae exhibit downregulation of Notch receptors notch1a and notch1b and show precocious differentiation of NeuroD-positive endocrine cells in the intrapancreatic duct and gut epithelium. Collectively, this study provides a novel insight into the development of pancreatic endocrine progenitors, revealing an essential role for sept7b in endocrine progenitor differentiation.
Collapse
Affiliation(s)
| | - Elina Hakonen
- Research Program for Molecular Neurology and Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland
| | - Jarkko Ustinov
- Research Program for Molecular Neurology and Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Research Program for Molecular Neurology and Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Sanna Lehtonen
- Department of Pathology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
94
|
Beer RL, Parsons MJ, Rovira M. Centroacinar cells: At the center of pancreas regeneration. Dev Biol 2016; 413:8-15. [PMID: 26963675 DOI: 10.1016/j.ydbio.2016.02.027] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/23/2016] [Accepted: 02/26/2016] [Indexed: 10/22/2022]
Abstract
The process of regeneration serves to heal injury by replacing missing cells. Understanding regeneration can help us replace cell populations lost during disease, such as the insulin-producing β cells lost in diabetic patients. Centroacinar cells (CACs) are a specialized ductal pancreatic cell type that act as progenitors to replace β cells in the zebrafish. However, whether CACs contribute to β-cell regeneration in adult mammals remains controversial. Here we review the current understanding of the role of CACs as endocrine progenitors during regeneration in zebrafish and mammals.
Collapse
Affiliation(s)
- Rebecca L Beer
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, United States.
| | - Michael J Parsons
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, United States; Department of Surgery, Johns Hopkins University, Baltimore, MD, United States
| | - Meritxell Rovira
- Genomic Programming of Beta-Cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
| |
Collapse
|
95
|
Kim HS, Lee MK. β-Cell regeneration through the transdifferentiation of pancreatic cells: Pancreatic progenitor cells in the pancreas. J Diabetes Investig 2016; 7:286-96. [PMID: 27330712 PMCID: PMC4847880 DOI: 10.1111/jdi.12475] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/27/2015] [Accepted: 01/04/2016] [Indexed: 12/17/2022] Open
Abstract
Pancreatic progenitor cell research has been in the spotlight, as these cells have the potential to replace pancreatic β‐cells for the treatment of type 1 and 2 diabetic patients with the absence or reduction of pancreatic β‐cells. During the past few decades, the successful treatment of diabetes through transplantation of the whole pancreas or isolated islets has nearly been achieved. However, novel sources of pancreatic islets or insulin‐producing cells are required to provide sufficient amounts of donor tissues. To overcome this limitation, the use of pancreatic progenitor cells is gaining more attention. In particular, pancreatic exocrine cells, such as duct epithelial cells and acinar cells, are attractive candidates for β‐cell regeneration because of their differentiation potential and pancreatic lineage characteristics. It has been assumed that β‐cell neogenesis from pancreatic progenitor cells could occur in pancreatic ducts in the postnatal stage. Several studies have shown that insulin‐producing cells can arise in the duct tissue of the adult pancreas. Acinar cells also might have the potential to differentiate into insulin‐producing cells. The present review summarizes recent progress in research on the transdifferentiation of pancreatic exocrine cells into insulin‐producing cells, especially duct and acinar cells.
Collapse
Affiliation(s)
- Hyo-Sup Kim
- Division of Endocrinology and Metabolism Department of Medicine Sungkyunkwan University School of Medicine Samsung Biomedical Research Institute Samsung Medical Center Seoul Korea
| | - Moon-Kyu Lee
- Division of Endocrinology and Metabolism Department of Medicine Sungkyunkwan University School of Medicine Samsung Biomedical Research Institute Samsung Medical Center Seoul Korea
| |
Collapse
|
96
|
Xu J, Cui J, Del Campo A, Shin CH. Four and a Half LIM Domains 1b (Fhl1b) Is Essential for Regulating the Liver versus Pancreas Fate Decision and for β-Cell Regeneration. PLoS Genet 2016; 12:e1005831. [PMID: 26845333 PMCID: PMC4741517 DOI: 10.1371/journal.pgen.1005831] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022] Open
Abstract
The liver and pancreas originate from overlapping embryonic regions, and single-cell lineage tracing in zebrafish has shown that Bone morphogenetic protein 2b (Bmp2b) signaling is essential for determining the fate of bipotential hepatopancreatic progenitors towards the liver or pancreas. Despite its pivotal role, the gene regulatory networks functioning downstream of Bmp2b signaling in this process are poorly understood. We have identified four and a half LIM domains 1b (fhl1b), which is primarily expressed in the prospective liver anlage, as a novel target of Bmp2b signaling. fhl1b depletion compromised liver specification and enhanced induction of pancreatic cells from endodermal progenitors. Conversely, overexpression of fhl1b favored liver specification and inhibited induction of pancreatic cells. By single-cell lineage tracing, we showed that fhl1b depletion led lateral endodermal cells, destined to become liver cells, to become pancreatic cells. Reversely, when fhl1b was overexpressed, medially located endodermal cells, fated to differentiate into pancreatic and intestinal cells, contributed to the liver by directly or indirectly modulating the discrete levels of pdx1 expression in endodermal progenitors. Moreover, loss of fhl1b increased the regenerative capacity of β-cells by increasing pdx1 and neurod expression in the hepatopancreatic ductal system. Altogether, these data reveal novel and critical functions of Fhl1b in the hepatic versus pancreatic fate decision and in β-cell regeneration. Lineage-specific multipotent progenitors play crucial roles in embryonic development, regeneration in adult tissues, and diseases such as cancer. Bone morphogenetic protein (Bmp) signaling is critical for regulating the cell fate choice of liver versus pancreas, two essential organs of body metabolism. Through transcriptome profiling of endodermal tissues exposed to increased or decreased Bmp2b signaling, we have discovered the zebrafish gene four and a half LIM domains 1b (fhl1b) as a novel target of Bmp2b signaling. fhl1b is primarily expressed in the prospective liver anlage. Loss- and gain-of-function analyses indicate that Fhl1b suppresses specification of the pancreas and induces the liver. By single-cell lineage tracing, we showed that depletion of fhl1b caused a liver-to-pancreas fate switch, while fhl1b overexpression redirected pancreatic progenitors to become liver cells. At later stages, Fhl1b regulates regeneration of insulin-secreting β-cells by directly or indirectly modulating pdx1 and neurod expression in the hepatopancreatic ductal system. Therefore, our work provides a novel paradigm of how Bmp signaling regulates the hepatic versus pancreatic fate decision and β-cell regeneration through its novel target Fhl1b.
Collapse
Affiliation(s)
- Jin Xu
- School of Biology and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Jiaxi Cui
- Max Planck Institute for Polymer Research, Mainz, Germany
| | | | - Chong Hyun Shin
- School of Biology and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
97
|
Jessen KR, Mirsky R, Arthur-Farraj P. The Role of Cell Plasticity in Tissue Repair: Adaptive Cellular Reprogramming. Dev Cell 2016; 34:613-20. [PMID: 26418293 DOI: 10.1016/j.devcel.2015.09.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/07/2015] [Accepted: 09/10/2015] [Indexed: 10/23/2022]
Abstract
It is becoming clear that a radical change of cell identity of differentiated cells in vivo, triggered by injury or other adversity, provides an essential route to recovery for many different mammalian tissues. This process, which we term adaptive cellular reprogramming, promotes regeneration in one of two ways: by providing a transient class of repair cells or by directly replacing cells lost during tissue damage. Controlling adaptive changes in cell fate in vivo in order to promote the body's own cell therapy, particularly by pharmacology rather than genetics, is likely to become an increasingly active area of future work.
Collapse
Affiliation(s)
- Kristjan R Jessen
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| | - Rhona Mirsky
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Peter Arthur-Farraj
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
98
|
Li M, Page-McCaw P, Chen W. FGF1 Mediates Overnutrition-Induced Compensatory β-Cell Differentiation. Diabetes 2016; 65:96-109. [PMID: 26420862 PMCID: PMC4686947 DOI: 10.2337/db15-0085] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 09/22/2015] [Indexed: 12/17/2022]
Abstract
Increased insulin demand resulting from insulin resistance and/or overnutrition induces a compensatory increase in β-cell mass. The physiological factors responsible for the compensation have not been fully characterized. In zebrafish, overnutrition rapidly induces compensatory β-cell differentiation through triggering the release of a paracrine signal from persistently activated β-cells. We identified Fgf1 signaling as a key component of the overnutrition-induced β-cell differentiation signal in a small molecule screen. Fgf1 was confirmed as the overnutrition-induced β-cell differentiation signal, as inactivation of fgf1 abolished the compensatory β-cell differentiation. Furthermore, expression of human FGF1 solely in β-cells in fgf1(-/-) animals rescued the compensatory response, indicating that β-cells can be the source of FGF1. Additionally, constitutive secretion of FGF1 with an exogenous signal peptide increased β-cell number in the absence of overnutrition. These results demonstrate that fgf1 is necessary and FGF1 expression in β-cells is sufficient for the compensatory β-cell differentiation. We further show that FGF1 is secreted during prolonged activation of cultured mammalian β-cells and that endoplasmic reticulum stress acts upstream of FGF1 release. Thus, the recently discovered antidiabetes function of FGF1 may act partially through increasing β-cell differentiation.
Collapse
Affiliation(s)
- Mingyu Li
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Patrick Page-McCaw
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Wenbiao Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
99
|
|
100
|
Ye L, Robertson MA, Mastracci TL, Anderson RM. An insulin signaling feedback loop regulates pancreas progenitor cell differentiation during islet development and regeneration. Dev Biol 2015; 409:354-69. [PMID: 26658317 DOI: 10.1016/j.ydbio.2015.12.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 11/12/2015] [Accepted: 12/02/2015] [Indexed: 02/06/2023]
Abstract
As one of the key nutrient sensors, insulin signaling plays an important role in integrating environmental energy cues with organism growth. In adult organisms, relative insufficiency of insulin signaling induces compensatory expansion of insulin-secreting pancreatic beta (β) cells. However, little is known about how insulin signaling feedback might influence neogenesis of β cells during embryonic development. Using genetic approaches and a unique cell transplantation system in developing zebrafish, we have uncovered a novel role for insulin signaling in the negative regulation of pancreatic progenitor cell differentiation. Blocking insulin signaling in the pancreatic progenitors hastened the expression of the essential β cell genes insulin and pdx1, and promoted β cell fate at the expense of alpha cell fate. In addition, loss of insulin signaling promoted β cell regeneration and destabilization of alpha cell character. These data indicate that insulin signaling constitutes a tunable mechanism for β cell compensatory plasticity during early development. Moreover, using a novel blastomere-to-larva transplantation strategy, we found that loss of insulin signaling in endoderm-committed blastomeres drove their differentiation into β cells. Furthermore, the extent of this differentiation was dependent on the function of the β cell mass in the host. Altogether, our results indicate that modulation of insulin signaling will be crucial for the development of β cell restoration therapies for diabetics; further clarification of the mechanisms of insulin signaling in β cell progenitors will reveal therapeutic targets for both in vivo and in vitro β cell generation.
Collapse
Affiliation(s)
- Lihua Ye
- Herman B Wells Center for Pediatric Research in the Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2043, Indianapolis, IN 46202, USA; Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2043, Indianapolis, IN 46202, USA
| | - Morgan A Robertson
- Herman B Wells Center for Pediatric Research in the Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2043, Indianapolis, IN 46202, USA
| | - Teresa L Mastracci
- Herman B Wells Center for Pediatric Research in the Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2043, Indianapolis, IN 46202, USA
| | - Ryan M Anderson
- Herman B Wells Center for Pediatric Research in the Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2043, Indianapolis, IN 46202, USA; Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2043, Indianapolis, IN 46202, USA.
| |
Collapse
|