1
|
Arias CF, Acosta FJ, Bertocchini F, Fernández-Arias C. A functional approach to homeostatic regulation. Biol Direct 2024; 19:134. [PMID: 39709473 DOI: 10.1186/s13062-024-00577-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/05/2024] [Indexed: 12/23/2024] Open
Abstract
In this work, we present a novel modeling framework for understanding the dynamics of homeostatic regulation. Inspired by engineering control theory, this framework incorporates unique features of biological systems. First, biological variables often play physiological roles, and taking this functional context into consideration is essential to fully understand the goals and constraints of homeostatic regulation. Second, biological signals are not abstract variables, but rather material molecules that may undergo complex turnover processes of synthesis and degradation. We suggest that the particular nature of biological signals may condition the type of information they can convey, and their potential role in shaping the dynamics and the ultimate purpose of homeostatic systems. We show that the dynamic interplay between regulated variables and control signals is a key determinant of biological homeostasis, challenging the necessity and the convenience of strictly extrapolating concepts from engineering control theory in modeling the dynamics of homeostatic systems. This work provides a simple, unified framework for studying biological regulation and identifies general principles that transcend molecular details of particular homeostatic mechanisms. We show how this approach can be naturally applied to apparently different regulatory systems, contributing to a deeper understanding of homeostasis as a fundamental process in living systems.
Collapse
Affiliation(s)
- Clemente F Arias
- Grupo Interdisciplinar de Sistemas Complejos de Madrid (GISC), 28040, Madrid, Spain.
| | - Francisco J Acosta
- Departamento de Ecología, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | | | - Cristina Fernández-Arias
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| |
Collapse
|
2
|
Krishnan G, Bagath M, Devaraj C, Soren NM. The signalling association of glucagon-like peptide-1 and its receptors in the gastrointestinal tract and GPR40 and insulin receptor in the pancreas of sheep. Gen Comp Endocrinol 2024; 358:114602. [PMID: 39226991 DOI: 10.1016/j.ygcen.2024.114602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/19/2024] [Accepted: 08/31/2024] [Indexed: 09/05/2024]
Abstract
The present study was aimed at gaining insight into the signalling relationship between glucagon-like peptide-1 (GLP-1) and its receptor (GLP-1R) in the regulation of glucose metabolism. Further, to assess the role of G-protein-coupled receptor 40 (GPR40) and insulin receptor (INSR) in the pancreas of sheep that were supplemented with calcium salts of long-chain fatty acids (CSFAs). An experiment was carried out over a period of 60 days with eighteen sheep, and they were fed with a standard basal diet. The sheep were divided into three groups: CSFA0 (without CSFAs), while CSFA3 and CSFA5 were supplemented with 3 % and 5 % of CSFAs, respectively. Plasma concentrations of GLP-1, insulin, glucagon, and glucose were assessed every two weeks. At the end of the experiment, sheep were slaughtered, and samples of gastrointestinal tract (GIT) epithelial tissues and pancreas were collected to assess the relative expression of mRNA of GPR40, GLP-1R, and INSR. Postprandial GLP-1 and insulin were increased by 3.7-4.1 and 1.45-1.5 times, respectively, in the CSFAs-supplemented groups compared to CSFA0. Post-feeding, glucagon and glucose levels decreased in CSFA3 and CSFA5 compared to CSFA0. The results indicated that the supplementation of LCFAs increased the expression of GLP-1R in the GIT and pancreas, as well as the mRNA of GPR40 and INSR in the pancreas. Chemosensing of LCFAs by GPR40 in the pancreas triggers signalling transduction, and enhanced GLP-1 and GLP-1R resulted in moderately increased insulin secretion and reduced glucagon levels. These combined effects, along with the glucose-lowering effect of GLP-1, effectively lowered glucose levels in normoglycemic sheep.
Collapse
Affiliation(s)
- G Krishnan
- Animal Physiology Division, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore 560030, India.
| | - M Bagath
- Animal Nutrition Division, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore 560030, India
| | - C Devaraj
- Bioenergetics and Environmental Sciences Division, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore 560030, India
| | - N M Soren
- Animal Nutrition Division, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore 560030, India
| |
Collapse
|
3
|
Bourgeois S, Coenen S, Degroote L, Willems L, Van Mulders A, Pierreux J, Heremans Y, De Leu N, Staels W. Harnessing beta cell regeneration biology for diabetes therapy. Trends Endocrinol Metab 2024; 35:951-966. [PMID: 38644094 DOI: 10.1016/j.tem.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 04/23/2024]
Abstract
The pandemic scale of diabetes mellitus is alarming, its complications remain devastating, and current treatments still pose a major burden on those affected and on the healthcare system as a whole. As the disease emanates from the destruction or dysfunction of insulin-producing pancreatic β-cells, a real cure requires their restoration and protection. An attractive strategy is to regenerate β-cells directly within the pancreas; however, while several approaches for β-cell regeneration have been proposed in the past, clinical translation has proven challenging. This review scrutinizes recent findings in β-cell regeneration and discusses their potential clinical implementation. Hereby, we aim to delineate a path for innovative, targeted therapies to help shift from 'caring for' to 'curing' diabetes.
Collapse
Affiliation(s)
- Stephanie Bourgeois
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Sophie Coenen
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Laure Degroote
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Lien Willems
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Annelore Van Mulders
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Julie Pierreux
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Yves Heremans
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Nico De Leu
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; Endocrinology, Universiteit Ziekenhuis Brussel (UZ Brussel), 1090 Brussels, Belgium; Endocrinology, ASZ Aalst, 9300 Aalst, Belgium.
| | - Willem Staels
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; Pediatric Endocrinology, Department of Pediatrics, KidZ Health Castle, Universiteit Ziekenhuis Brussel (UZ Brussel), 1090 Brussels, Belgium.
| |
Collapse
|
4
|
Jiang M, Wang N, Zhang Y, Zhang J, Li Y, Yan X, Zhang H, Li C, Guan Y, Liang B, Zhang W, Wu Y. Insulin receptor isoform B is required for efficient proinsulin processing in pancreatic β cells. iScience 2024; 27:110017. [PMID: 39021804 PMCID: PMC11253548 DOI: 10.1016/j.isci.2024.110017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/27/2024] [Accepted: 05/14/2024] [Indexed: 07/20/2024] Open
Abstract
The insulin receptor (INSR, IR) has two isoforms, IRA and IRB, through alternative splicing. However, their distinct functions in vivo remain unclear. Here we generated β cell-specific IRB knockout (KO) mice (βIRBKO). The KO mice displayed worsened hyperinsulinemia and hyperproinsulinemia in diet-induced obesity due to impaired proinsulin processing in β cells. Mechanistically, loss of IRB suppresses eukaryotic translation initiation factor 4G1 (eIF4G1) by stabilizing the transcriptional receptor sterol-regulatory element binding protein 1 (SREBP1). Moreover, excessive autocrine proinsulin in βIRBKO mice enhances the activity of extracellular signal-regulated kinase (ERK) through the remaining IRA to further stabilize nuclear SREBP1, forming a feedback loop. Collectively, our study paves the way to dissecting the isoform-specific function of IR in vivo and highlights the important roles of IRB in insulin processing and protecting β cells from lipotoxicity in obesity.
Collapse
Affiliation(s)
- Mingchao Jiang
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning 116000, China
| | - Ning Wang
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning 116000, China
| | - Yuqin Zhang
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning 116000, China
| | - Jinjin Zhang
- Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250021, China
| | - Youwei Li
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning 116000, China
- Haidu College, Qingdao Agricultural University, Laiyang, Shandong 265200, China
| | - Xiu Yan
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning 116000, China
| | - Honghao Zhang
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning 116000, China
| | - Chengbin Li
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Bin Liang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China
| | - Weiping Zhang
- Department of Pathophysiology, Naval Medical University, Shanghai 200433, China
| | - Yingjie Wu
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning 116000, China
- Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250021, China
| |
Collapse
|
5
|
Murata N, Nishimura K, Harada N, Kitakaze T, Yoshihara E, Inui H, Yamaji R. Insulin reduces endoplasmic reticulum stress-induced apoptosis by decreasing mitochondrial hyperpolarization and caspase-12 in INS-1 pancreatic β-cells. Physiol Rep 2024; 12:e16106. [PMID: 38884322 PMCID: PMC11181300 DOI: 10.14814/phy2.16106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
Pancreatic β-cell mass is a critical determinant of insulin secretion. Severe endoplasmic reticulum (ER) stress causes β-cell apoptosis; however, the mechanisms of progression and suppression are not yet fully understood. Here, we report that the autocrine/paracrine function of insulin reduces ER stress-induced β-cell apoptosis. Insulin reduced the ER-stress inducer tunicamycin- and thapsigargin-induced cell viability loss due to apoptosis in INS-1 β-cells. Moreover, the effect of insulin was greater than that of insulin-like growth factor-1 at physiologically relevant concentrations. Insulin did not attenuate the ER stress-induced increase in unfolded protein response genes. ER stress did not induce cytochrome c release from mitochondria. Mitochondrial hyperpolarization was induced by ER stress and prevented by insulin. The protonophore/mitochondrial oxidative phosphorylation uncoupler, but not the antioxidants N-acetylcysteine and α-tocopherol, exhibited potential cytoprotection during ER stress. Both procaspase-12 and cleaved caspase-12 levels increased under ER stress. The caspase-12 inhibitor Z-ATAD-FMK decreased ER stress-induced apoptosis. Caspase-12 overexpression reduced cell viability, which was diminished in the presence of insulin. Insulin decreased caspase-12 levels at the post-translational stages. These results demonstrate that insulin protects against ER stress-induced β-cell apoptosis in this cell line. Furthermore, mitochondrial hyperpolarization and increased caspase-12 levels are involved in ER stress-induced and insulin-suppressed β-cell apoptosis.
Collapse
Affiliation(s)
- Nanako Murata
- Department of Applied Biological Chemistry, Graduate School of AgricultureOsaka Metropolitan UniversitySakaiOsakaJapan
| | - Kana Nishimura
- Division of Applied Life Sciences, Graduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
| | - Naoki Harada
- Department of Applied Biological Chemistry, Graduate School of AgricultureOsaka Metropolitan UniversitySakaiOsakaJapan
- Division of Applied Life Sciences, Graduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
| | - Tomoya Kitakaze
- Department of Applied Biological Chemistry, Graduate School of AgricultureOsaka Metropolitan UniversitySakaiOsakaJapan
- Division of Applied Life Sciences, Graduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
| | - Eiji Yoshihara
- The Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCaliforniaUSA
- David Geffen School of Medicine at University of California Los AngelesLos AngelesCaliforniaUSA
| | - Hiroshi Inui
- Department of Applied Biological Chemistry, Graduate School of AgricultureOsaka Metropolitan UniversitySakaiOsakaJapan
- Division of Applied Life Sciences, Graduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
- Department of Health and NutritionOtemae UniversityOsakaJapan
| | - Ryoichi Yamaji
- Department of Applied Biological Chemistry, Graduate School of AgricultureOsaka Metropolitan UniversitySakaiOsakaJapan
- Division of Applied Life Sciences, Graduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
- Center for Research and Development of BioresourcesOsaka Metropolitan UniversitySakaiOsakaJapan
| |
Collapse
|
6
|
Mittendorfer B, Johnson JD, Solinas G, Jansson PA. Insulin Hypersecretion as Promoter of Body Fat Gain and Hyperglycemia. Diabetes 2024; 73:837-843. [PMID: 38768368 PMCID: PMC11109786 DOI: 10.2337/dbi23-0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/26/2024] [Indexed: 05/22/2024]
Affiliation(s)
- Bettina Mittendorfer
- Departments of Medicine and Nutrition & Exercise Physiology, School of Medicine, University of Missouri, Columbia, MO
| | - James D. Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Giovanni Solinas
- Department of Molecular and Clinical Medicine, School of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Per-Anders Jansson
- Department of Molecular and Clinical Medicine, School of Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
7
|
Koca SB, Kulali MA, Göğüş B, Demirbilek H. Type A insulin resistance syndrome due to a novel heterozygous c.3486_3503del (p.Arg1163_Ala1168del) INSR gene mutation in an adolescent girl and her mother. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e210305. [PMID: 38289143 PMCID: PMC10948035 DOI: 10.20945/2359-4292-2021-0305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 12/05/2022] [Indexed: 02/01/2024]
Abstract
Mutations in the insulin receptor (INSR) gene may present with variable clinical phenotypes. We report herein a novel heterozygous INSR mutation in an adolescent girl with type A insulin resistance syndrome and her mother.The index case was a 12-year-old girl without obesity who presented with excessive hair growth, especially in the chest and back area, and hyperpigmentation on the back of the neck (acanthosis nigricans). Acanthosis nigricans was first observed at the age of 11 years. On physical examination, the patient had acanthosis nigricans and hypertrichosis with no acne. Systolic and diastolic blood pressure measurement was within the normal range for age and sex. Laboratory tests revealed fasting hyperglycemia, fasting and postprandial hyperinsulinemia, elevated HbA1c level, and biochemical hyperandrogenemia. Fasting plasma lipids were normal. A diagnosis of type A insulin resistance syndrome was considered, and INSR gene mutation analysis was performed. Next generation sequence analysis was performed with the use of primers containing exon/exon-intron junctions in the INSR gene, and a novel heterozygous c.3486_3503delGAGAAACTGCATGGTCGC/p.Arg1163_Ala1168del change was detected in exon 19 of the INSR gene. In segregation analysis, the same variant was detected in the patient's mother, who had a milder clinical phenotype.We reported a novel, heterozygous, p.Arg1163_Ala1168del mutation in exon 19 of the INSR gene in a patient with type A insulin resistance syndrome, expanding the mutation database. The same mutation was associated with variable phenotypical severity in two subjects within the same family.
Collapse
Affiliation(s)
- Serkan Bilge Koca
- Afyonkarahisar Health Sciences University, Faculty of Medicine, Department of Pediatrics, Division of Pediatric Endocrinology, Afyonkarahisar, Turkey,
| | - Melike Ataseven Kulali
- Afyonkarahisar Health Sciences University, Faculty of Medicine, Department of Pediatrics, Division of Pediatric Genetics, Afyonkarahisar, Turkey
| | - Başak Göğüş
- Afyonkarahisar Health Sciences University, Faculty of Medicine, Department of Medical Genetics, Afyonkarahisar, Turkey
| | - Hüseyin Demirbilek
- Hacettepe University, Faculty of Medicine, Department of Pediatrics, Division of Pediatric Endocrinology, Ankara, Turkey
| |
Collapse
|
8
|
Cota P, Caliskan ÖS, Bastidas-Ponce A, Jing C, Jaki J, Saber L, Czarnecki O, Taskin D, Blöchinger AK, Kurth T, Sterr M, Burtscher I, Krahmer N, Lickert H, Bakhti M. Insulin regulates human pancreatic endocrine cell differentiation in vitro. Mol Metab 2024; 79:101853. [PMID: 38103636 PMCID: PMC10765254 DOI: 10.1016/j.molmet.2023.101853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/21/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
OBJECTIVE The consequences of mutations in genes associated with monogenic forms of diabetes on human pancreas development cannot be studied in a time-resolved fashion in vivo. More specifically, if recessive mutations in the insulin gene influence human pancreatic endocrine lineage formation is still an unresolved question. METHODS To model the extremely reduced insulin levels in patients with recessive insulin gene mutations, we generated a novel knock-in H2B-Cherry reporter human induced pluripotent stem cell (iPSC) line expressing no insulin upon differentiation to stem cell-derived (SC-) β cells in vitro. Differentiation of iPSCs into the pancreatic and endocrine lineage, combined with immunostaining, Western blotting and proteomics analysis phenotypically characterized the insulin gene deficiency in SC-islets. Furthermore, we leveraged FACS analysis and confocal microscopy to explore the impact of insulin shortage on human endocrine cell induction, composition, differentiation and proliferation. RESULTS Interestingly, insulin-deficient SC-islets exhibited low insulin receptor (IR) signaling when stimulated with glucose but displayed increased IR sensitivity upon treatment with exogenous insulin. Furthermore, insulin shortage did not alter neurogenin-3 (NGN3)-mediated endocrine lineage induction. Nevertheless, lack of insulin skewed the SC-islet cell composition with an increased number in SC-β cell formation at the expense of SC-α cells. Finally, insulin deficiency reduced the rate of SC-β cell proliferation but had no impact on the expansion of SC-α cells. CONCLUSIONS Using iPSC disease modelling, we provide first evidence of insulin function in human pancreatic endocrine lineage formation. These findings help to better understand the phenotypic impact of recessive insulin gene mutations during pancreas development and shed light on insulin gene function beside its physiological role in blood glucose regulation.
Collapse
Affiliation(s)
- Perla Cota
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Özüm Sehnaz Caliskan
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute of Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Changying Jing
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Munich medical research school (MMRS), Ludwig Maximilian University (LMU), Munich, Germany
| | - Jessica Jaki
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Lama Saber
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Oliver Czarnecki
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Damla Taskin
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
| | - Anna Karolina Blöchinger
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Thomas Kurth
- Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform Core Facility Electron Microscopy and Histology, Technische Universität Dresden, Dresden, Germany
| | - Michael Sterr
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Natalie Krahmer
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute of Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany.
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
9
|
Zhang Z, Li M, Sun T, Zhang Z, Liu C. FOXM1: Functional Roles of FOXM1 in Non-Malignant Diseases. Biomolecules 2023; 13:biom13050857. [PMID: 37238726 DOI: 10.3390/biom13050857] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Forkhead box (FOX) proteins are a wing-like helix family of transcription factors in the DNA-binding region. By mediating the activation and inhibition of transcription and interactions with all kinds of transcriptional co-regulators (MuvB complexes, STAT3, β-catenin, etc.), they play significant roles in carbohydrate and fat metabolism, biological aging and immune regulation, development, and diseases in mammals. Recent studies have focused on translating these essential findings into clinical applications in order to improve quality of life, investigating areas such as diabetes, inflammation, and pulmonary fibrosis, and increase human lifespan. Early studies have shown that forkhead box M1 (FOXM1) functions as a key gene in pathological processes in multiple diseases by regulating genes related to proliferation, the cell cycle, migration, and apoptosis and genes related to diagnosis, therapy, and injury repair. Although FOXM1 has long been studied in relation to human diseases, its role needs to be elaborated on. FOXM1 expression is involved in the development or repair of multiple diseases, including pulmonary fibrosis, pneumonia, diabetes, liver injury repair, adrenal lesions, vascular diseases, brain diseases, arthritis, myasthenia gravis, and psoriasis. The complex mechanisms involve multiple signaling pathways, such as WNT/β-catenin, STAT3/FOXM1/GLUT1, c-Myc/FOXM1, FOXM1/SIRT4/NF-κB, and FOXM1/SEMA3C/NRP2/Hedgehog. This paper reviews the key roles and functions of FOXM1 in kidney, vascular, lung, brain, bone, heart, skin, and blood vessel diseases to elucidate the role of FOXM1 in the development and progression of human non-malignant diseases and makes suggestions for further research.
Collapse
Affiliation(s)
- Zhenwang Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Mengxi Li
- School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning 437100, China
| | - Tian Sun
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
- Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Zhengrong Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
- Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Chao Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
- Medical Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| |
Collapse
|
10
|
Bhat N, Mani A. Dysregulation of Lipid and Glucose Metabolism in Nonalcoholic Fatty Liver Disease. Nutrients 2023; 15:2323. [PMID: 37242206 PMCID: PMC10222271 DOI: 10.3390/nu15102323] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/08/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) is a highly prevalent condition affecting approximately a quarter of the global population. It is associated with increased morbidity, mortality, economic burden, and healthcare costs. The disease is characterized by the accumulation of lipids in the liver, known as steatosis, which can progress to more severe stages such as steatohepatitis, fibrosis, cirrhosis, and even hepatocellular carcinoma (HCC). This review focuses on the mechanisms that contribute to the development of diet-induced steatosis in an insulin-resistant liver. Specifically, it discusses the existing literature on carbon flux through glycolysis, ketogenesis, TCA (Tricarboxylic Acid Cycle), and fatty acid synthesis pathways in NAFLD, as well as the altered canonical insulin signaling and genetic predispositions that lead to the accumulation of diet-induced hepatic fat. Finally, the review discusses the current therapeutic efforts that aim to ameliorate various pathologies associated with NAFLD.
Collapse
Affiliation(s)
| | - Arya Mani
- Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA
| |
Collapse
|
11
|
Moroni-González D, Sarmiento-Ortega VE, Diaz A, Brambila E, Treviño S. Pancreas-Liver-Adipose Axis: Target of Environmental Cadmium Exposure Linked to Metabolic Diseases. TOXICS 2023; 11:223. [PMID: 36976988 PMCID: PMC10059892 DOI: 10.3390/toxics11030223] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 06/18/2023]
Abstract
Cadmium has been well recognized as a critical toxic agent in acute and chronic poisoning cases in occupational and nonoccupational settings and environmental exposure situations. Cadmium is released into the environment after natural and anthropogenic activities, particularly in contaminated and industrial areas, causing food pollution. In the body, cadmium has no biological activity, but it accumulates primarily in the liver and kidney, which are considered the main targets of its toxicity, through oxidative stress and inflammation. However, in the last few years, this metal has been linked to metabolic diseases. The pancreas-liver-adipose axis is largely affected by cadmium accumulation. Therefore, this review aims to collect bibliographic information that establishes the basis for understanding the molecular and cellular mechanisms linked to cadmium with carbohydrate, lipids, and endocrine impairments that contribute to developing insulin resistance, metabolic syndrome, prediabetes, and diabetes.
Collapse
Affiliation(s)
- Diana Moroni-González
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, Meritorious Autonomous University of Puebla, Ciudad Universitaria, Puebla 72560, Mexico
| | - Victor Enrique Sarmiento-Ortega
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, Meritorious Autonomous University of Puebla, Ciudad Universitaria, Puebla 72560, Mexico
| | - Alfonso Diaz
- Department of Pharmacy, Faculty of Chemistry Science, Meritorious Autonomous University of Puebla, 22 South. FCQ9, Ciudad Universitaria, Puebla 72560, Mexico
| | - Eduardo Brambila
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, Meritorious Autonomous University of Puebla, Ciudad Universitaria, Puebla 72560, Mexico
| | - Samuel Treviño
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, Meritorious Autonomous University of Puebla, Ciudad Universitaria, Puebla 72560, Mexico
| |
Collapse
|
12
|
Castell AL, Goubault C, Ethier M, Fergusson G, Tremblay C, Baltz M, Dal Soglio D, Ghislain J, Poitout V. β Cell mass expansion during puberty involves serotonin signaling and determines glucose homeostasis in adulthood. JCI Insight 2022; 7:160854. [PMID: 36107617 PMCID: PMC9675460 DOI: 10.1172/jci.insight.160854] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 09/14/2022] [Indexed: 01/07/2023] Open
Abstract
Puberty is associated with transient insulin resistance that normally recedes at the end of puberty; however, in overweight children, insulin resistance persists, leading to an increased risk of type 2 diabetes. The mechanisms whereby pancreatic β cells adapt to pubertal insulin resistance, and how they are affected by the metabolic status, have not been investigated. Here, we show that puberty is associated with a transient increase in β cell proliferation in rats and humans of both sexes. In rats, β cell proliferation correlated with a rise in growth hormone (GH) levels. Serum from pubertal rats and humans promoted β cell proliferation, suggesting the implication of a circulating factor. In pubertal rat islets, expression of genes of the GH/serotonin (5-hydroxytryptamine [5-HT]) pathway underwent changes consistent with a proliferative effect. Inhibition of the pro-proliferative 5-HT receptor isoform HTR2B blocked the increase in β cell proliferation in pubertal islets ex vivo and in vivo. Peripubertal metabolic stress blunted β cell proliferation during puberty and led to altered glucose homeostasis later in life. This study identifies a role of GH/GH receptor/5-HT/HTR2B signaling in the control of β cell mass expansion during puberty and identifies a mechanistic link between pubertal obesity and the risk of developing type 2 diabetes.
Collapse
Affiliation(s)
- Anne-Laure Castell
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Medicine and
| | - Clara Goubault
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Pharmacology and Physiology, University of Montreal, Quebec, Canada
| | - Mélanie Ethier
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Grace Fergusson
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Caroline Tremblay
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Marie Baltz
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Dorothée Dal Soglio
- CHU Sainte-Justine, Montreal, Quebec, Canada.,Department of Pathology and Cell Biology, University of Montreal, Montreal, Quebec, Canada
| | - Julien Ghislain
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Medicine and
| |
Collapse
|
13
|
Sharma S, Behl T, Sehgal A, Singh S, Sharma N, Bhatia S, Al-Harassi A, Bungau S, Mostafavi E. Possible Role of Wnt Signaling Pathway in Diabetic Retinopathy. Curr Drug Targets 2022; 23:1372-1380. [PMID: 35232336 DOI: 10.2174/1389450123666220301110140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/08/2021] [Accepted: 12/30/2021] [Indexed: 01/25/2023]
Abstract
The core of impaired vision in working people suffering from insulin-dependent and noninsulin- dependent diabetes mellitus is diabetic retinopathy (DR). The Wnt Protein Ligands family influences various processes; this ensures the cells are able to interact and co-ordinate various mobile functions, including cell growth, division, survival, apoptosis, migration, and cell destiny. The extracellular Wnt signal activates other signals. It is seen that Wnt pathways play an important role in inflammation, oxidative stress, and angiogenesis. It has been illustrated that the canonically preserved Wnt signaling system has a vital role in the homeostasis of adulthood. Developmental disorders in each of these stages will lead to serious eye problems and eventually blindness. There is, therefore, a need to specifically organize and regulate the growth of ocular tissues. In tissue specification and polarities, axonal exhaust, and maintenance of cells, especially in the central nervous system, Wnt/frizzled pathways play an important role. Thus, Wnt route antagonists may act as have been possible therapeutic options in DR by inhibiting aberrant Wnt signals. Elaborative and continued research in this area will help in the advancement of current knowledge in the field of DR, and eventually, this can lead to the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Sheetu Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Bidholi, Dehradun, India
| | - Aayush Sehgal
- GHG Khalsa College of Pharmacy, Gurusar Sadhar, Ludhiana, Punjab, India
| | - Sukhbir Singh
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Neelam Sharma
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Ahmed Al-Harassi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
14
|
Shirakawa J, Togashi Y, Basile G, Okuyama T, Inoue R, Fernandez M, Kyohara M, De Jesus DF, Goto N, Zhang W, Tsuno T, Kin T, Pan H, Dreyfuss JM, Shapiro AJ, Yi P, Terauchi Y, Kulkarni RN. E2F1 transcription factor mediates a link between fat and islets to promote β cell proliferation in response to acute insulin resistance. Cell Rep 2022; 41:111436. [PMID: 36198264 PMCID: PMC9617565 DOI: 10.1016/j.celrep.2022.111436] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/21/2022] [Accepted: 09/08/2022] [Indexed: 02/03/2023] Open
Abstract
Prevention or amelioration of declining β cell mass is a potential strategy to cure diabetes. Here, we report the pathways utilized by β cells to robustly replicate in response to acute insulin resistance induced by S961, a pharmacological insulin receptor antagonist. Interestingly, pathways that include CENP-A and the transcription factor E2F1 that are independent of insulin signaling and its substrates appeared to mediate S961-induced β cell multiplication. Consistently, pharmacological inhibition of E2F1 blocks β-cell proliferation in S961-injected mice. Serum from S961-treated mice recapitulates replication of β cells in mouse and human islets in an E2F1-dependent manner. Co-culture of islets with adipocytes isolated from S961-treated mice enables β cells to duplicate, while E2F1 inhibition limits their growth even in the presence of adipocytes. These data suggest insulin resistance-induced proliferative signals from adipocytes activate E2F1, a potential therapeutic target, to promote β cell compensation.
Collapse
Affiliation(s)
- Jun Shirakawa
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA,Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 3718512, Japan,Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama 2360004, Japan,Correspondence: (J.S.), (R.N.K.)
| | - Yu Togashi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama 2360004, Japan
| | - Giorgio Basile
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Tomoko Okuyama
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama 2360004, Japan
| | - Ryota Inoue
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 3718512, Japan
| | - Megan Fernandez
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Mayu Kyohara
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama 2360004, Japan
| | - Dario F. De Jesus
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Nozomi Goto
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama 2360004, Japan
| | - Wei Zhang
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Takahiro Tsuno
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 3718512, Japan
| | - Tatsuya Kin
- Clinical Islet Laboratory and Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Hui Pan
- Bioinformatics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jonathan M. Dreyfuss
- Bioinformatics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - A.M. James Shapiro
- Clinical Islet Laboratory and Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Peng Yi
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama 2360004, Japan
| | - Rohit N. Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA,Lead contact,Correspondence: (J.S.), (R.N.K.)
| |
Collapse
|
15
|
Brasil BB, Masaji S, Martins BT, Jiang H, Song N, Athena A S, Lucas B, François M, Wei-Jun Q, Rohit KN, Ronald KC. Apolipoprotein C3 and circulating mediators of preadipocyte proliferation in states of lipodystrophy. Mol Metab 2022; 64:101572. [PMID: 35964946 PMCID: PMC9418991 DOI: 10.1016/j.molmet.2022.101572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 02/02/2023] Open
Abstract
Adipogenesis is a complex process controlled by intrinsic and extrinsic factors that regulate preadipocyte proliferation, adipogenic capacity and maturation of metabolic function. Here we show that insulin and IGF-1 receptors are essential for mature adipocyte survival and that deletion of both IR and IGF1R specifically in fat using a tamoxifen inducible-AdipoQ-Cre (Ai-DKO) leads to rapid and severe loss of adipocytes in all depots, associated with a metabolic syndrome characterized by hypertriglyceridemia, hyperglycemia, hyperinsulinemia, fatty liver, and pancreatic beta cell proliferation. In this model, this pathological phenotype reverses over a few weeks, in large part, due to preadipocyte proliferation and adipose tissue regeneration. Incubation of preadipocytes with serum from the Ai-DKO mice in vitro stimulates cell proliferation, and this effect can be mimicked by conditioned media from liver slices of Ai-DKO mice, but not by media of cultured Ai-DKO adipocytes, indicating a hepatic origin of the growth factor. Proteomic analysis of serum reveals apolipoprotein C3 (APOC3), a protein secreted by liver, as one of the most upregulated proteins in the Ai-DKO mice. In vitro, purified and delipidated APOC3 stimulates preadipocyte proliferation, however, knockdown of hepatic APOC3 in vivo in Ai-DKO mice is not sufficient to block adipose regeneration. Thus, lipodystrophy is associated with presence of increased preadipocyte-stimulating growth factors in serum. Our study indicates that APOC3 is one contributing factor to preadipocyte proliferation, however, other still-unidentified circulating growth factors are also likely present in Ai-DKO mice. Identification of these factors may provide a new approach to regulation of adipose mass in health and disease.
Collapse
Affiliation(s)
- Brandao Bruna Brasil
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Sakaguchi Masaji
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA; Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Batista Thiago Martins
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Hu Jiang
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Dept. of Medicine, BIDMC, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Nie Song
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Schepmoes Athena A
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | | | - Moreau François
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Qian Wei-Jun
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Kulkarni N Rohit
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Dept. of Medicine, BIDMC, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Kahn C Ronald
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Jain C, Bilekova S, Lickert H. Targeting pancreatic β cells for diabetes treatment. Nat Metab 2022; 4:1097-1108. [PMID: 36131204 DOI: 10.1038/s42255-022-00618-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 07/13/2022] [Indexed: 11/09/2022]
Abstract
Insulin is a life-saving drug for patients with type 1 diabetes; however, even today, no pharmacotherapy can prevent the loss or dysfunction of pancreatic insulin-producing β cells to stop or reverse disease progression. Thus, pancreatic β cells have been a main focus for cell-replacement and regenerative therapies as a curative treatment for diabetes. In this Review, we highlight recent advances toward the development of diabetes therapies that target β cells to enhance proliferation, redifferentiation and protection from cell death and/or enable selective killing of senescent β cells. We describe currently available therapies and their mode of action, as well as insufficiencies of glucagon-like peptide 1 (GLP-1) and insulin therapies. We discuss and summarize data collected over the last decades that support the notion that pharmacological targeting of β cell insulin signalling might protect and/or regenerate β cells as an improved treatment of patients with diabetes.
Collapse
Affiliation(s)
- Chirag Jain
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Immunology Discovery, Genentech Inc., South San Francisco, CA, USA
| | - Sara Bilekova
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Chair of β-Cell Biology, Technische Universität München, School of Medicine, Klinikum Rechts der Isar, München, Germany.
| |
Collapse
|
17
|
Lv C, Sun Y, Zhang ZY, Aboelela Z, Qiu X, Meng ZX. β-cell dynamics in type 2 diabetes and in dietary and exercise interventions. J Mol Cell Biol 2022; 14:6656373. [PMID: 35929791 PMCID: PMC9710517 DOI: 10.1093/jmcb/mjac046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/07/2022] [Accepted: 08/03/2022] [Indexed: 01/14/2023] Open
Abstract
Pancreatic β-cell dysfunction and insulin resistance are two of the major causes of type 2 diabetes (T2D). Recent clinical and experimental studies have suggested that the functional capacity of β-cells, particularly in the first phase of insulin secretion, is a primary contributor to the progression of T2D and its associated complications. Pancreatic β-cells undergo dynamic compensation and decompensation processes during the development of T2D, in which metabolic stresses such as endoplasmic reticulum stress, oxidative stress, and inflammatory signals are key regulators of β-cell dynamics. Dietary and exercise interventions have been shown to be effective approaches for the treatment of obesity and T2D, especially in the early stages. Whilst the targeted tissues and underlying mechanisms of dietary and exercise interventions remain somewhat vague, accumulating evidence has implicated the improvement of β-cell functional capacity. In this review, we summarize recent advances in the understanding of the dynamic adaptations of β-cell function in T2D progression and clarify the effects and mechanisms of dietary and exercise interventions on β-cell dysfunction in T2D. This review provides molecular insights into the therapeutic effects of dietary and exercise interventions on T2D, and more importantly, it paves the way for future research on the related underlying mechanisms for developing precision prevention and treatment of T2D.
Collapse
Affiliation(s)
- Chengan Lv
- Department of Pathology and Pathophysiology and Metabolic Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yuchen Sun
- Department of Pathology and Pathophysiology and Metabolic Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China,Zhejiang University–University of Edinburgh Institute (ZJE), Zhejiang University, Haining 314400, China
| | - Zhe Yu Zhang
- Department of Pathology and Pathophysiology and Metabolic Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zeyad Aboelela
- Department of Pathology and Pathophysiology and Metabolic Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China,Bachelors of Surgery, Bachelors of Medicine (MBBS), Zhejiang University School of Medicine, Hangzhou 310003, China
| | | | | |
Collapse
|
18
|
Khan D, Moffett RC, Flatt PR, Tarasov AI. Classical and non-classical islet peptides in the control of β-cell function. Peptides 2022; 150:170715. [PMID: 34958851 DOI: 10.1016/j.peptides.2021.170715] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/25/2021] [Accepted: 12/17/2021] [Indexed: 12/25/2022]
Abstract
The dual role of the pancreas as both an endocrine and exocrine gland is vital for food digestion and control of nutrient metabolism. The exocrine pancreas secretes enzymes into the small intestine aiding digestion of sugars and fats, whereas the endocrine pancreas secretes a cocktail of hormones into the blood, which is responsible for blood glucose control and regulation of carbohydrate, protein and fat metabolism. Classical islet hormones, insulin, glucagon, pancreatic polypeptide and somatostatin, interact in an autocrine and paracrine manner, to fine-tube the islet function and insulin secretion to the needs of the body. Recently pancreatic islets have been reported to express a number of non-classical peptide hormones involved in metabolic signalling, whose major production site was believed to reside outside pancreas, e.g. in the small intestine. We highlight the key non-classical islet peptides, and consider their involvement, together with established islet hormones, in regulation of stimulus-secretion coupling as well as proliferation, survival and transdifferentiation of β-cells. We furthermore focus on the paracrine interaction between classical and non-classical islet hormones in the maintenance of β-cell function. Understanding the functional relationships between these islet peptides might help to develop novel, more efficient treatments for diabetes and related metabolic disorders.
Collapse
Affiliation(s)
- Dawood Khan
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK.
| | - R Charlotte Moffett
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Peter R Flatt
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Andrei I Tarasov
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| |
Collapse
|
19
|
Skovsø S, Panzhinskiy E, Kolic J, Cen HH, Dionne DA, Dai XQ, Sharma RB, Elghazi L, Ellis CE, Faulkner K, Marcil SAM, Overby P, Noursadeghi N, Hutchinson D, Hu X, Li H, Modi H, Wildi JS, Botezelli JD, Noh HL, Suk S, Gablaski B, Bautista A, Kim R, Cras-Méneur C, Flibotte S, Sinha S, Luciani DS, Nislow C, Rideout EJ, Cytrynbaum EN, Kim JK, Bernal-Mizrachi E, Alonso LC, MacDonald PE, Johnson JD. Beta-cell specific Insr deletion promotes insulin hypersecretion and improves glucose tolerance prior to global insulin resistance. Nat Commun 2022; 13:735. [PMID: 35136059 PMCID: PMC8826929 DOI: 10.1038/s41467-022-28039-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 01/05/2022] [Indexed: 01/23/2023] Open
Abstract
Insulin receptor (Insr) protein is present at higher levels in pancreatic β-cells than in most other tissues, but the consequences of β-cell insulin resistance remain enigmatic. Here, we use an Ins1cre knock-in allele to delete Insr specifically in β-cells of both female and male mice. We compare experimental mice to Ins1cre-containing littermate controls at multiple ages and on multiple diets. RNA-seq of purified recombined β-cells reveals transcriptomic consequences of Insr loss, which differ between female and male mice. Action potential and calcium oscillation frequencies are increased in Insr knockout β-cells from female, but not male mice, whereas only male βInsrKO islets have reduced ATP-coupled oxygen consumption rate and reduced expression of genes involved in ATP synthesis. Female βInsrKO and βInsrHET mice exhibit elevated insulin release in ex vivo perifusion experiments, during hyperglycemic clamps, and following i.p. glucose challenge. Deletion of Insr does not alter β-cell area up to 9 months of age, nor does it impair hyperglycemia-induced proliferation. Based on our data, we adapt a mathematical model to include β-cell insulin resistance, which predicts that β-cell Insr knockout improves glucose tolerance depending on the degree of whole-body insulin resistance. Indeed, glucose tolerance is significantly improved in female βInsrKO and βInsrHET mice compared to controls at 9, 21 and 39 weeks, and also in insulin-sensitive 4-week old males. We observe no improved glucose tolerance in older male mice or in high fat diet-fed mice, corroborating the prediction that global insulin resistance obscures the effects of β-cell specific insulin resistance. The propensity for hyperinsulinemia is associated with mildly reduced fasting glucose and increased body weight. We further validate our main in vivo findings using an Ins1-CreERT transgenic line and find that male mice have improved glucose tolerance 4 weeks after tamoxifen-mediated Insr deletion. Collectively, our data show that β-cell insulin resistance in the form of reduced β-cell Insr contributes to hyperinsulinemia in the context of glucose stimulation, thereby improving glucose homeostasis in otherwise insulin sensitive sex, dietary and age contexts.
Collapse
Affiliation(s)
- Søs Skovsø
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Evgeniy Panzhinskiy
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jelena Kolic
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Haoning Howard Cen
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Derek A Dionne
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Xiao-Qing Dai
- Alberta Diabetes Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada
| | - Rohit B Sharma
- Division of Endocrinology, Diabetes and Metabolism and the Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
| | - Lynda Elghazi
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI, USA
| | - Cara E Ellis
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Katharine Faulkner
- Department of Mathematics, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie A M Marcil
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Peter Overby
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Nilou Noursadeghi
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Daria Hutchinson
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Xiaoke Hu
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Hong Li
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Honey Modi
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jennifer S Wildi
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - J Diego Botezelli
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Hye Lim Noh
- Program in Molecular Medicine University of Massachusetts Medical School, Worcester, MA, USA
- Charles River Laboratories, Shrewsbury, MA, USA
| | - Sujin Suk
- Program in Molecular Medicine University of Massachusetts Medical School, Worcester, MA, USA
| | - Brian Gablaski
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
- Charles River Laboratories, Shrewsbury, MA, USA
| | - Austin Bautista
- Alberta Diabetes Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada
| | - Ryekjang Kim
- Alberta Diabetes Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada
| | - Corentin Cras-Méneur
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Stephane Flibotte
- UBC Life Sciences Institute Bioinformatics Facility, University of British Columbia, Vancouver, BC, Canada
| | - Sunita Sinha
- UBC Sequencing and Bioinformatics Consortium, Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Dan S Luciani
- BC Children's Hospital Research Institute, Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Corey Nislow
- UBC Sequencing and Bioinformatics Consortium, Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth J Rideout
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Eric N Cytrynbaum
- Department of Mathematics, University of British Columbia, Vancouver, BC, Canada
| | - Jason K Kim
- Program in Molecular Medicine University of Massachusetts Medical School, Worcester, MA, USA
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Ernesto Bernal-Mizrachi
- Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine and Miami VA Health Care System, Miami, FL, USA
| | - Laura C Alonso
- Division of Endocrinology, Diabetes and Metabolism and the Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
| | - Patrick E MacDonald
- Alberta Diabetes Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada
| | - James D Johnson
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
20
|
Moullé VS. Autonomic control of pancreatic beta cells: What is known on the regulation of insulin secretion and beta-cell proliferation in rodents and humans. Peptides 2022; 148:170709. [PMID: 34896576 DOI: 10.1016/j.peptides.2021.170709] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/17/2021] [Accepted: 12/07/2021] [Indexed: 11/21/2022]
Abstract
Insulin secretion and pancreatic beta-cell proliferation are tightly regulated by several signals such as hormones, nutrients, and neurotransmitters. However, the autonomic control of beta cells is not fully understood. In this review, we describe mechanisms involved in insulin secretion as well as metabolic and mitogenic actions on its target tissues. Since pancreatic islets are physically connected to the brain by nerves, parasympathetic and sympathetic neurotransmitters can directly potentiate or repress insulin secretion and beta-cell proliferation. Finally, we highlight the role of the autonomic nervous system in metabolic diseases such as diabetes and obesity.
Collapse
|
21
|
Dietary Protein Modulates the Efficacy of Taurine Supplementation on Adaptive Islet Function and Morphology in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1370:267-278. [DOI: 10.1007/978-3-030-93337-1_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
22
|
Norouzi M, Saghi H, Mohebbati R, Mirzavi F, Afshari AR, Soukhtanloo M. Effects of some anti-diabetic herbal extracts on the insulin-degrading enzyme in human colon cancer Caco-2 cell line. AVICENNA JOURNAL OF PHYTOMEDICINE 2022; 12:548-558. [PMID: 36249459 PMCID: PMC9516401 DOI: 10.22038/ajp.2022.19982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/24/2022] [Accepted: 02/14/2022] [Indexed: 12/02/2022]
Abstract
OBJECTIVE Type 2 diabetes mellitus (T2DM) is a condition characterized by insufficient insulin production or insulin resistance. The insulin-degrading enzyme (IDE) is responsible for degrading insulin and is a potential drug target for T2DM treatment. Numerous activities have been proposed for plant extracts, but research on the effects of plant extracts on IDE expression and activity is riddled with drawbacks. MATERIALS AND METHODS We investigated the effect of Phaseolus vulgaris, Allium cepa, Portulaca oleracea, Cinnamomum verum, and Citrullus colocynthis extracts on the expression and activity of IDE in the Caco-2 cell line. RESULTS Findings of RT-PCR showed that IDE gene expression was reduced following treatment with P. vulgaris, C. colocynthis, and C. verum extracts. The results of IDE activity with fluorogenic peptide substrate V also indicated that P. vulgaris, C. colocynthis, and P. oleracea extracts reduced IDE activity in a significant and dose-dependent manner. CONCLUSION The hydroalcoholic extracts studied, except for A. cepa, can prevent insulin degradation by reducing the expression and activity of the IDE enzyme. This new insight into the effects of herbal medicines on IDE activity can help future studies.
Collapse
Affiliation(s)
- Mahtab Norouzi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Saghi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Mohebbati
- Department of Physiology, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran,Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farshad Mirzavi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Reza Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran,Pharmacological Research Center of Medicinal Plants, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran,Corresponding Author: Tel: +98-5138002366, Fax: +98-5138828574,
| |
Collapse
|
23
|
Sánchez-Archidona AR, Cruciani-Guglielmacci C, Roujeau C, Wigger L, Lallement J, Denom J, Barovic M, Kassis N, Mehl F, Weitz J, Distler M, Klose C, Simons K, Ibberson M, Solimena M, Magnan C, Thorens B. Plasma triacylglycerols are biomarkers of β-cell function in mice and humans. Mol Metab 2021; 54:101355. [PMID: 34634522 PMCID: PMC8602044 DOI: 10.1016/j.molmet.2021.101355] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/27/2021] [Accepted: 10/06/2021] [Indexed: 12/13/2022] Open
Abstract
Objectives To find plasma biomarkers prognostic of type 2 diabetes, which could also inform on pancreatic β-cell deregulations or defects in the function of insulin target tissues. Methods We conducted a systems biology approach to characterize the plasma lipidomes of C57Bl/6J, DBA/2J, and BALB/cJ mice under different nutritional conditions, as well as their pancreatic islet and liver transcriptomes. We searched for correlations between plasma lipids and tissue gene expression modules. Results We identified strong correlation between plasma triacylglycerols (TAGs) and islet gene modules that comprise key regulators of glucose- and lipid-regulated insulin secretion and of the insulin signaling pathway, the two top hits were Gck and Abhd6 for negative and positive correlations, respectively. Correlations were also found between sphingomyelins and islet gene modules that overlapped in part with the gene modules correlated with TAGs. In the liver, the gene module most strongly correlated with plasma TAGs was enriched in mRNAs encoding fatty acid and carnitine transporters as well as multiple enzymes of the β-oxidation pathway. In humans, plasma TAGs also correlated with the expression of several of the same key regulators of insulin secretion and the insulin signaling pathway identified in mice. This cross-species comparative analysis further led to the identification of PITPNC1 as a candidate regulator of glucose-stimulated insulin secretion. Conclusion TAGs emerge as biomarkers of a liver-to-β-cell axis that links hepatic β-oxidation to β-cell functional mass and insulin secretion. Plasma triacylglycerols correlated with genes controlling β-cell mass and function. Plasma triacylglycerols correlated with genes controlling liver β-oxidation. In humans, triacylglycerols also correlated with key regulators of insulin secretion. Mouse and human data identified PITPNC1 as a candidate regulator of insulin secretion. Triacylglycerols are biomarkers of the liver-to-β-cell axis and β-cell function.
Collapse
Affiliation(s)
- Ana Rodríguez Sánchez-Archidona
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; Vital-IT Group, SIB Swiss Institute for Bioinformatics, 1015 Lausanne, Switzerland.
| | | | - Clara Roujeau
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Leonore Wigger
- Vital-IT Group, SIB Swiss Institute for Bioinformatics, 1015 Lausanne, Switzerland.
| | | | - Jessica Denom
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France.
| | - Marko Barovic
- Department of Molecular Diabetology, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany.
| | - Nadim Kassis
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France.
| | - Florence Mehl
- Vital-IT Group, SIB Swiss Institute for Bioinformatics, 1015 Lausanne, Switzerland.
| | - Jurgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital, TU Dresden, Dresden, Germany.
| | - Marius Distler
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital, TU Dresden, Dresden, Germany.
| | | | | | - Mark Ibberson
- Vital-IT Group, SIB Swiss Institute for Bioinformatics, 1015 Lausanne, Switzerland.
| | - Michele Solimena
- Department of Molecular Diabetology, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany.
| | | | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
24
|
Ramzy A, Kieffer TJ. Altered islet prohormone processing: A cause or consequence of diabetes? Physiol Rev 2021; 102:155-208. [PMID: 34280055 DOI: 10.1152/physrev.00008.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Peptide hormones are first produced as larger precursor prohormones that require endoproteolytic cleavage to liberate the mature hormones. A structurally conserved but functionally distinct family of nine prohormone convertase enzymes (PCs) are responsible for cleavage of protein precursors of which PC1/3 and PC2 are known to be exclusive to neuroendocrine cells and responsible for prohormone cleavage. Differential expression of PCs within tissues define prohormone processing; whereas glucagon is the major product liberated from proglucagon via PC2 in pancreatic α-cells, proglucagon is preferentially processed by PC1/3 in intestinal L cells to produce glucagon-like peptides 1 and 2 (GLP-1, GLP-2). Beyond our understanding of processing of islet prohormones in healthy islets, there is convincing evidence that proinsulin, proIAPP, and proglucagon processing is altered during prediabetes and diabetes. There is predictive value of elevated circulating proinsulin or proinsulin : C-peptide ratio for progression to type 2 diabetes and elevated proinsulin or proinsulin : C-peptide is predictive for development of type 1 diabetes in at risk groups. After onset of diabetes, patients have elevated circulating proinsulin and proIAPP and proinsulin may be an autoantigen in type 1 diabetes. Further, preclinical studies reveal that α-cells have altered proglucagon processing during diabetes leading to increased GLP-1 production. We conclude that despite strong associative data, current evidence is inconclusive on the potential causal role of impaired prohormone processing in diabetes, and suggest that future work should focus on resolving the question of whether altered prohormone processing is a causal driver or merely a consequence of diabetes pathology.
Collapse
Affiliation(s)
- Adam Ramzy
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Timothy J Kieffer
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
25
|
The Roles of the IGF Axis in the Regulation of the Metabolism: Interaction and Difference between Insulin Receptor Signaling and IGF-I Receptor Signaling. Int J Mol Sci 2021; 22:ijms22136817. [PMID: 34202916 PMCID: PMC8268872 DOI: 10.3390/ijms22136817] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/24/2022] Open
Abstract
It has been well established that insulin-like growth factors (IGFs) mainly mediate long-term actions in cell fates, whereas insulin predominantly exerts its role on metabolic activity. Indeed, insulin mediates multiple anabolic biological activities in glucose and amino acid transport, lipid and protein synthesis, the induction of glycogen, the inhibition of gluconeogenesis, lipolysis, and protein degradation. The interactions and differences between insulin receptor signaling and IGF-I receptor signaling in the metabolism and the cell fates are quite complicated. Because of the overlapping actions of IGF-I singling with insulin signaling, it has been difficult to distinguish the role of both signaling mechanisms on the metabolism. Furthermore, comprehensive information on the IGF-I function in respective tissues remains insufficient. Therefore, we need to clarify the precise roles of IGF-I signaling on the metabolism separate from those of insulin signaling. This review focuses on the metabolic roles of IGFs in the respective tissues, especially in terms of comparison with those of insulin, by overviewing the metabolic phenotypes of tissue-specific IGF-I and insulin receptor knockout mice, as well as those in mice treated with the dual insulin receptor/IGF-I receptor inhibitor OSI-906.
Collapse
|
26
|
Coppola I, Brouwers B, Meulemans S, Ramos-Molina B, Creemers JWM. Differential Effects of Furin Deficiency on Insulin Receptor Processing and Glucose Control in Liver and Pancreatic β Cells of Mice. Int J Mol Sci 2021; 22:6344. [PMID: 34198511 PMCID: PMC8231939 DOI: 10.3390/ijms22126344] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 01/04/2023] Open
Abstract
The insulin receptor (IR) is critically involved in maintaining glucose homeostasis. It undergoes proteolytic cleavage by proprotein convertases, which is an essential step for its activation. The importance of the insulin receptor in liver is well established, but its role in pancreatic β cells is still controversial. In this study, we investigated the cleavage of the IR by the proprotein convertase FURIN in β cells and hepatocytes, and the contribution of the IR in pancreatic β cells and liver to glucose homeostasis. β-cell-specific Furin knockout (βFurKO) mice were glucose intolerant, but liver-specific Furin knockout (LFurKO) mice were normoglycemic. Processing of the IR was blocked in βFurKO cells, but unaffected in LFurKO mice. Most strikingly, glucose homeostasis in β-cell-specific IR knockout (βIRKO) mice was normal in younger mice (up to 20 weeks), and only mildly affected in older mice (24 weeks). In conclusion, FURIN cleaves the IR non-redundantly in β cells, but redundantly in liver. Furthermore, we demonstrated that the IR in β cells plays a limited role in glucose homeostasis.
Collapse
Affiliation(s)
- Ilaria Coppola
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; (I.C.); (B.B.); (S.M.)
| | - Bas Brouwers
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; (I.C.); (B.B.); (S.M.)
| | - Sandra Meulemans
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; (I.C.); (B.B.); (S.M.)
| | - Bruno Ramos-Molina
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; (I.C.); (B.B.); (S.M.)
- Obesity and Metabolism Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain
| | - John W. M. Creemers
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; (I.C.); (B.B.); (S.M.)
| |
Collapse
|
27
|
Maachi H, Ghislain J, Tremblay C, Poitout V. Pronounced proliferation of non-beta cells in response to beta-cell mitogens in isolated human islets of Langerhans. Sci Rep 2021; 11:11283. [PMID: 34050242 PMCID: PMC8163757 DOI: 10.1038/s41598-021-90643-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 05/06/2021] [Indexed: 11/09/2022] Open
Abstract
The potential to treat diabetes by increasing beta-cell mass is driving a major effort to identify beta-cell mitogens. Demonstration of mitogen activity in human beta cells is frequently performed in ex vivo assays. However, reported disparities in the efficacy of beta-cell mitogens led us to investigate the sources of this variability. We studied 35 male (23) and female (12) human islet batches covering a range of donor ages and BMI. Islets were kept intact or dispersed into single cells and cultured in the presence of harmine, glucose, or heparin-binding epidermal growth factor-like growth factor (HB-EGF), and subsequently analyzed by immunohistochemistry or flow cytometry. Proliferating cells were identified by double labeling with EdU and Ki67 and glucagon, c-peptide or Nkx6.1, and cytokeratin-19 to respectively label alpha, beta, and ductal cells. Harmine and HB-EGF stimulated human beta-cell proliferation, but the effect of glucose was dependent on the assay and the donor. Harmine potently stimulated alpha-cell proliferation and both harmine and HB-EGF increased proliferation of insulin- and glucagon-negative cells, including cytokeratin 19-positive cells. Given the abundance of non-beta cells in human islet preparations, our results suggest that assessment of beta-cell mitogens requires complementary approaches and rigorous identification of cell identity using multiple markers.
Collapse
Affiliation(s)
- Hasna Maachi
- Montreal Diabetes Research Center, CRCHUM, 900 rue St Denis, Montreal, QC, H2X 0A9, Canada.,Department of Pharmacology and Physiology, University of Montreal, Montreal, QC, Canada
| | - Julien Ghislain
- Montreal Diabetes Research Center, CRCHUM, 900 rue St Denis, Montreal, QC, H2X 0A9, Canada
| | - Caroline Tremblay
- Montreal Diabetes Research Center, CRCHUM, 900 rue St Denis, Montreal, QC, H2X 0A9, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, CRCHUM, 900 rue St Denis, Montreal, QC, H2X 0A9, Canada. .,Department of Medicine, University of Montreal, Montreal, QC, Canada.
| |
Collapse
|
28
|
Klein KR, Walker CP, McFerren AL, Huffman H, Frohlich F, Buse JB. Carbohydrate Intake Prior to Oral Glucose Tolerance Testing. J Endocr Soc 2021; 5:bvab049. [PMID: 33928207 PMCID: PMC8059359 DOI: 10.1210/jendso/bvab049] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Indexed: 12/19/2022] Open
Abstract
With the emergence of glycated hemoglobin as a diagnostic test for diabetes, oral glucose tolerance tests (OGTTs) have become rare in endocrinology practice. As they have moved out of favor, the importance of patient instructions on preparation prior to OGTT has faded from memory. Decades-old literature, well-known to endocrinologists a generation ago, emphasized the importance of carbohydrate intake prior to OGTT. In this expert endocrine consult, we discuss an OGTT performed in a research setting without adequate carbohydrate intake at the evening meal prior to the OGTT. The resultant elevated plasma glucose levels at 1-hour and 2-hours mimicked the loss of first-phase insulin release seen in early type 1 and type 2 diabetes. With clinical concern that the research participant had evolving type 1 or type 2 diabetes, the volunteer was subjected to additional testing and experienced anxiety. Repeat OGTT was normal after adequate carbohydrate intake (>150 grams/day and >50 grams the evening prior to overnight fast for the study). The physiology of this phenomenon is explored and is likely mediated through beta cell adaptation and alteration in peripheral glucose uptake in response to nutrient exposure. The learnings of decades ago have clearly faded, and this literature should be revisited to ensure that OGTT results are not compromised when ordered for clinical or research purposes.
Collapse
Affiliation(s)
- Klara R Klein
- Division of Endocrinology and Metabolism, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Christopher P Walker
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Center for Neurostimulation, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Amber L McFerren
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Center for Neurostimulation, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Halie Huffman
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Flavio Frohlich
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Center for Neurostimulation, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John B Buse
- Division of Endocrinology and Metabolism, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
29
|
Takatani T, Shirakawa J, Shibue K, Gupta MK, Kim H, Lu S, Hu J, White MF, Kennedy RT, Kulkarni RN. Insulin receptor substrate 1, but not IRS2, plays a dominant role in regulating pancreatic alpha cell function in mice. J Biol Chem 2021; 296:100646. [PMID: 33839150 PMCID: PMC8131928 DOI: 10.1016/j.jbc.2021.100646] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/24/2021] [Accepted: 04/07/2021] [Indexed: 11/29/2022] Open
Abstract
Dysregulated glucagon secretion deteriorates glycemic control in type 1 and type 2 diabetes. Although insulin is known to regulate glucagon secretion via its cognate receptor (insulin receptor, INSR) in pancreatic alpha cells, the role of downstream proteins and signaling pathways underlying insulin's activities are not fully defined. Using in vivo (knockout) and in vitro (knockdown) studies targeting insulin receptor substrate (IRS) proteins, we compared the relative roles of IRS1 and IRS2 in regulating alpha cell function. Alpha cell-specific IRS1-knockout mice exhibited glucose intolerance and inappropriate glucagon suppression during glucose tolerance tests. In contrast, alpha cell-specific IRS2-knockout animals manifested normal glucose tolerance and suppression of glucagon secretion after glucose administration. Alpha cell lines with stable IRS1 knockdown could not repress glucagon mRNA expression and exhibited a reduction in phosphorylation of AKT Ser/Thr kinase (AKT, at Ser-473 and Thr-308). AlphaIRS1KD cells also displayed suppressed global protein translation, including reduced glucagon expression, impaired cytoplasmic Ca2+ response, and mitochondrial dysfunction. This was supported by the identification of novel IRS1-specific downstream target genes, Trpc3 and Cartpt, that are associated with glucagon regulation in alpha cells. These results provide evidence that IRS1, rather than IRS2, is a dominant regulator of pancreatic alpha cell function.
Collapse
Affiliation(s)
- Tomozumi Takatani
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jun Shirakawa
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Gunma, Japan
| | - Kimitaka Shibue
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Manoj K Gupta
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Cell Therapy Translational Engine (CTTE), Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Hyunki Kim
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shusheng Lu
- Departments of Chemistry and Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jiang Hu
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Morris F White
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert T Kennedy
- Departments of Chemistry and Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
30
|
Taki K, Takagi H, Hirose T, Sun R, Yaginuma H, Mizoguchi A, Kobayashi T, Sugiyama M, Tsunekawa T, Onoue T, Hagiwara D, Ito Y, Iwama S, Suga H, Banno R, Sakano D, Kume S, Arima H. Dietary sodium chloride attenuates increased β-cell mass to cause glucose intolerance in mice under a high-fat diet. PLoS One 2021; 16:e0248065. [PMID: 33730054 PMCID: PMC7968668 DOI: 10.1371/journal.pone.0248065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/19/2021] [Indexed: 11/19/2022] Open
Abstract
Excessive sodium salt (NaCl) or fat intake is associated with a variety of increased health risks. However, whether excessive NaCl intake accompanied by a high-fat diet (HFD) affects glucose metabolism has not been elucidated. In this study, C57BL/6J male mice were fed a normal chow diet (NCD), a NCD plus high-NaCl diet (NCD plus NaCl), a HFD, or a HFD plus high-NaCl diet (HFD plus NaCl) for 30 weeks. No significant differences in body weight gain, insulin sensitivity, and glucose tolerance were observed between NCD-fed and NCD plus NaCl-fed mice. In contrast, body and liver weights were decreased, but the weight of epididymal white adipose tissue was increased in HFD plus NaCl-fed compared to HFD-fed mice. HFD plus NaCl-fed mice had lower plasma glucose levels in an insulin tolerance test, and showed higher plasma glucose and lower plasma insulin levels in an intraperitoneal glucose tolerance test compared to HFD-fed mice. The β-cell area and number of islets were decreased in HFD plus NaCl-fed compared to HFD-fed mice. Increased Ki67-positive β-cells, and increased expression levels of Ki67, CyclinB1, and CyclinD1 mRNA in islets were observed in HFD-fed but not HFD plus NaCl-fed mice when compared to NCD-fed mice. Our data suggest that excessive NaCl intake accompanied by a HFD exacerbates glucose intolerance, with impairment in insulin secretion caused by the attenuation of expansion of β-cell mass in the pancreas.
Collapse
Affiliation(s)
- Keigo Taki
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Hiroshi Takagi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
- * E-mail:
| | - Tomonori Hirose
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Runan Sun
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Hiroshi Yaginuma
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Akira Mizoguchi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Tomoko Kobayashi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Mariko Sugiyama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Taku Tsunekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Takeshi Onoue
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Daisuke Hagiwara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Yoshihiro Ito
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Ryoichi Banno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
- Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya, Japan
| | - Daisuke Sakano
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Midori-ku, Yokohama, Kanagawa, Japan
| | - Shoen Kume
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Midori-ku, Yokohama, Kanagawa, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| |
Collapse
|
31
|
Lithovius V, Saarimäki-Vire J, Balboa D, Ibrahim H, Montaser H, Barsby T, Otonkoski T. SUR1-mutant iPS cell-derived islets recapitulate the pathophysiology of congenital hyperinsulinism. Diabetologia 2021; 64:630-640. [PMID: 33404684 DOI: 10.1007/s00125-020-05346-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/13/2020] [Indexed: 12/27/2022]
Abstract
AIMS/HYPOTHESIS Congenital hyperinsulinism caused by mutations in the KATP-channel-encoding genes (KATPHI) is a potentially life-threatening disorder of the pancreatic beta cells. No optimal medical treatment is available for patients with diazoxide-unresponsive diffuse KATPHI. Therefore, we aimed to create a model of KATPHI using patient induced pluripotent stem cell (iPSC)-derived islets. METHODS We derived iPSCs from a patient carrying a homozygous ABCC8V187D mutation, which inactivates the sulfonylurea receptor 1 (SUR1) subunit of the KATP-channel. CRISPR-Cas9 mutation-corrected iPSCs were used as controls. Both were differentiated to stem cell-derived islet-like clusters (SC-islets) and implanted into NOD-SCID gamma mice. RESULTS SUR1-mutant and -corrected iPSC lines both differentiated towards the endocrine lineage, but SUR1-mutant stem cells generated 32% more beta-like cells (SC-beta cells) (64.6% vs 49.0%, p = 0.02) and 26% fewer alpha-like cells (16.1% vs 21.8% p = 0.01). SUR1-mutant SC-beta cells were 61% more proliferative (1.23% vs 0.76%, p = 0.006), and this phenotype could be induced in SUR1-corrected cells with pharmacological KATP-channel inactivation. The SUR1-mutant SC-islets secreted 3.2-fold more insulin in low glucose conditions (0.0174% vs 0.0054%/min, p = 0.0021) and did not respond to KATP-channel-acting drugs in vitro. Mice carrying grafts of SUR1-mutant SC-islets presented with 38% lower fasting blood glucose (4.8 vs 7.7 mmol/l, p = 0.009) and their grafts failed to efficiently shut down insulin secretion during induced hypoglycaemia. Explanted SUR1-mutant grafts displayed an increase in SC-beta cell proportion and SC-beta cell nucleomegaly, which was independent of proliferation. CONCLUSIONS/INTERPRETATION We have created a model recapitulating the known pathophysiology of KATPHI both in vitro and in vivo. We have also identified a novel role for KATP-channel activity during human islet development. This model will enable further studies for the improved understanding and clinical management of KATPHI without the need for primary patient tissue.
Collapse
Affiliation(s)
- Väinö Lithovius
- Stem Cells and Metabolism Research Program in the Faculty of Medicine of the University of Helsinki, Helsinki, Finland.
| | - Jonna Saarimäki-Vire
- Stem Cells and Metabolism Research Program in the Faculty of Medicine of the University of Helsinki, Helsinki, Finland
| | - Diego Balboa
- Stem Cells and Metabolism Research Program in the Faculty of Medicine of the University of Helsinki, Helsinki, Finland
- Bioinformatics and Genomics Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Hazem Ibrahim
- Stem Cells and Metabolism Research Program in the Faculty of Medicine of the University of Helsinki, Helsinki, Finland
| | - Hossam Montaser
- Stem Cells and Metabolism Research Program in the Faculty of Medicine of the University of Helsinki, Helsinki, Finland
| | - Tom Barsby
- Stem Cells and Metabolism Research Program in the Faculty of Medicine of the University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Stem Cells and Metabolism Research Program in the Faculty of Medicine of the University of Helsinki, Helsinki, Finland.
| |
Collapse
|
32
|
Inceptor counteracts insulin signalling in β-cells to control glycaemia. Nature 2021; 590:326-331. [DOI: 10.1038/s41586-021-03225-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022]
|
33
|
Diwekar-Joshi M, Watve M. Driver versus navigator causation in biology: the case of insulin and fasting glucose. PeerJ 2020; 8:e10396. [PMID: 33365205 PMCID: PMC7735078 DOI: 10.7717/peerj.10396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/29/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND In biomedicine, inferring causal relation from experimental intervention or perturbation is believed to be a more reliable approach than inferring causation from cross-sectional correlation. However, we point out here that even in interventional inference there are logical traps. In homeostatic systems, causality in a steady state can be qualitatively different from that in a perturbed state. On a broader scale there is a need to differentiate driver causality from navigator causality. A driver is essential for reaching a destination but may not have any role in deciding the destination. A navigator on the other hand has a role in deciding the destination and the path but may not be able to drive the system to the destination. The failure to differentiate between types of causalities is likely to have resulted into many misinterpretations in physiology and biomedicine. METHODS We illustrate this by critically re-examining a specific case of the causal role of insulin in glucose homeostasis using five different approaches (1) Systematic review of tissue specific insulin receptor knock-outs, (2) Systematic review of insulin suppression and insulin enhancement experiments, (3) Differentiating steady state and post-meal state glucose levels in streptozotocin treated rats in primary experiments, (4) Mathematical and theoretical considerations and (5) Glucose-insulin relationship in human epidemiological data. RESULTS All the approaches converge on the inference that although insulin action hastens the return to a steady state after a glucose load, there is no evidence that insulin action determines the steady state level of glucose. Insulin, unlike the popular belief in medicine, appears to be a driver but not a navigator for steady state glucose level. It is quite likely therefore that the current line of clinical action in the field of type 2 diabetes has limited success largely because it is based on a misinterpretation of glucose-insulin relationship. The insulin-glucose example suggests that we may have to carefully re-examine causal inferences from perturbation experiments and set up revised norms for experimental design for causal inference.
Collapse
Affiliation(s)
- Manawa Diwekar-Joshi
- Biology, Indian Institute of Science Education and Research, Pune, Maharashtra, India
| | - Milind Watve
- Deenanath Mangeshkar Hospital and Research Centre, Pune, Maharashtra, India
| |
Collapse
|
34
|
Arous C, Mizgier ML, Rickenbach K, Pinget M, Bouzakri K, Wehrle-Haller B. Integrin and autocrine IGF2 pathways control fasting insulin secretion in β-cells. J Biol Chem 2020; 295:16510-16528. [PMID: 32934005 PMCID: PMC7864053 DOI: 10.1074/jbc.ra120.012957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/09/2020] [Indexed: 12/20/2022] Open
Abstract
Elevated levels of fasting insulin release and insufficient glucose-stimulated insulin secretion (GSIS) are hallmarks of diabetes. Studies have established cross-talk between integrin signaling and insulin activity, but more details of how integrin-dependent signaling impacts the pathophysiology of diabetes are needed. Here, we dissected integrin-dependent signaling pathways involved in the regulation of insulin secretion in β-cells and studied their link to the still debated autocrine regulation of insulin secretion by insulin/insulin-like growth factor (IGF) 2-AKT signaling. We observed for the first time a cooperation between different AKT isoforms and focal adhesion kinase (FAK)-dependent adhesion signaling, which either controlled GSIS or prevented insulin secretion under fasting conditions. Indeed, β-cells form integrin-containing adhesions, which provide anchorage to the pancreatic extracellular matrix and are the origin of intracellular signaling via FAK and paxillin. Under low-glucose conditions, β-cells adopt a starved adhesion phenotype consisting of actin stress fibers and large peripheral focal adhesion. In contrast, glucose stimulation induces cell spreading, actin remodeling, and point-like adhesions that contain phospho-FAK and phosphopaxillin, located in small protrusions. Rat primary β-cells and mouse insulinomas showed an adhesion remodeling during GSIS resulting from autocrine insulin/IGF2 and AKT1 signaling. However, under starving conditions, the maintenance of stress fibers and the large adhesion phenotype required autocrine IGF2-IGF1 receptor signaling mediated by AKT2 and elevated FAK-kinase activity and ROCK-RhoA levels but low levels of paxillin phosphorylation. This starved adhesion phenotype prevented excessive insulin granule release to maintain low insulin secretion during fasting. Thus, deregulation of the IGF2 and adhesion-mediated signaling may explain dysfunctions observed in diabetes.
Collapse
Affiliation(s)
- Caroline Arous
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland.
| | - Maria Luisa Mizgier
- UMR DIATHEC, Centre Européen d'Etude du Diabète, UMR DIATHEC, Strasbourg, France
| | - Katharina Rickenbach
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland
| | - Michel Pinget
- UMR DIATHEC, Centre Européen d'Etude du Diabète, UMR DIATHEC, Strasbourg, France
| | - Karim Bouzakri
- UMR DIATHEC, Centre Européen d'Etude du Diabète, UMR DIATHEC, Strasbourg, France
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland
| |
Collapse
|
35
|
Abstract
The current paradigm of type 2 diabetes (T2D) is gluco-centric, being exclusively categorized by glycemic characteristics. The gluco-centric paradigm views hyperglycemia as the primary target, being driven by resistance to insulin combined with progressive beta cells failure, and considers glycemic control its ultimate treatment goal. Most importantly, the gluco-centric paradigm considers the non-glycemic diseases associated with T2D, e.g., obesity, dyslipidemia, hypertension, macrovascular disease, microvascular disease and fatty liver as 'risk factors' and/or 'outcomes' and/or 'comorbidities', rather than primary inherent disease aspects of T2D. That is in spite of their high prevalence (60-90%) and major role in profiling T2D morbidity and mortality. Moreover, the gluco-centric paradigm fails to realize that the non-glycemic diseases of T2D are driven by insulin and, except for glycemic control, response to insulin in T2D is essentially the rule rather than the exception. Failure of the gluco-centric paradigm to offer an exhaustive unifying view of the glycemic and non-glycemic diseases of T2D may have contributed to T2D being still an unmet need. An mTORC1-centric paradigm maintains that hyperactive mTORC1 drives the glycemic and non-glycemic disease aspects of T2D. Hyperactive mTORC1 is proposed to act as double-edged agent, namely, to interfere with glycemic control by disrupting the insulin receptor-Akt transduction pathway, while concomitantly driving the non-glycemic diseases of T2D. The mTORC1-centric paradigm may offer a novel perspective for T2D in terms of pathogenesis, clinical focus and treatment strategy.
Collapse
Affiliation(s)
- Jacob Bar-Tana
- Hebrew University Medical School, 91120, Jerusalem, Israel.
| |
Collapse
|
36
|
Delgadillo-Puga C, Noriega LG, Morales-Romero AM, Nieto-Camacho A, Granados-Portillo O, Rodríguez-López LA, Alemán G, Furuzawa-Carballeda J, Tovar AR, Cisneros-Zevallos L, Torre-Villalvazo I. Goat's Milk Intake Prevents Obesity, Hepatic Steatosis and Insulin Resistance in Mice Fed A High-Fat Diet by Reducing Inflammatory Markers and Increasing Energy Expenditure and Mitochondrial Content in Skeletal Muscle. Int J Mol Sci 2020; 21:ijms21155530. [PMID: 32752280 PMCID: PMC7432599 DOI: 10.3390/ijms21155530] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/22/2020] [Accepted: 07/29/2020] [Indexed: 12/26/2022] Open
Abstract
Goat's milk is a rich source of bioactive compounds (peptides, conjugated linoleic acid, short chain fatty acids, monounsaturated and polyunsaturated fatty acids, polyphenols such as phytoestrogens and minerals among others) that exert important health benefits. However, goat's milk composition depends on the type of food provided to the animal and thus, the abundance of bioactive compounds in milk depends on the dietary sources of the goat feed. The metabolic impact of goat milk rich in bioactive compounds during metabolic challenges such as a high-fat (HF) diet has not been explored. Thus, we evaluated the effect of milk from goats fed a conventional diet, a conventional diet supplemented with 30% Acacia farnesiana (AF) pods or grazing on metabolic alterations in mice fed a HF diet. Interestingly, the incorporation of goat's milk in the diet decreased body weight and body fat mass, improved glucose tolerance, prevented adipose tissue hypertrophy and hepatic steatosis in mice fed a HF diet. These effects were associated with an increase in energy expenditure, augmented oxidative fibers in skeletal muscle, and reduced inflammatory markers. Consequently, goat's milk can be considered a non-pharmacologic strategy to improve the metabolic alterations induced by a HF diet. Using the body surface area normalization method gave a conversion equivalent daily human intake dose of 1.4 to 2.8 glasses (250 mL per glass/day) of fresh goat milk for an adult of 60 kg, which can be used as reference for future clinical studies.
Collapse
Affiliation(s)
- Claudia Delgadillo-Puga
- Departamento de Nutrición Animal Dr. Fernando Pérez-Gil Romo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico
- Correspondence: (C.D.-P.); (I.T.-V.); Tel.: +52-55-54870900 (C.D.-P. & I.T.-V.)
| | - Lilia G. Noriega
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico; (L.G.N.); (O.G.-P.); (L.A.R.-L.); (G.A.); (A.R.T.)
| | - Aurora M. Morales-Romero
- Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de Mexico 04510, Mexico;
| | - Antonio Nieto-Camacho
- Instituto de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de Mexico 04510, Mexico;
| | - Omar Granados-Portillo
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico; (L.G.N.); (O.G.-P.); (L.A.R.-L.); (G.A.); (A.R.T.)
| | - Leonardo A. Rodríguez-López
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico; (L.G.N.); (O.G.-P.); (L.A.R.-L.); (G.A.); (A.R.T.)
| | - Gabriela Alemán
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico; (L.G.N.); (O.G.-P.); (L.A.R.-L.); (G.A.); (A.R.T.)
| | - Janette Furuzawa-Carballeda
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico;
| | - Armando R. Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico; (L.G.N.); (O.G.-P.); (L.A.R.-L.); (G.A.); (A.R.T.)
| | - Luis Cisneros-Zevallos
- Department of Horticultural Sciences, Texas A&M University, College Station, TX 77843-2133, USA;
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Ivan Torre-Villalvazo
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Ciudad de Mexico 14080, Mexico; (L.G.N.); (O.G.-P.); (L.A.R.-L.); (G.A.); (A.R.T.)
- Correspondence: (C.D.-P.); (I.T.-V.); Tel.: +52-55-54870900 (C.D.-P. & I.T.-V.)
| |
Collapse
|
37
|
Zhao LL, Liu HL, Luo S, Walsh KM, Li W, Wei Q. Associations of novel variants in PIK3C3, INSR and MAP3K4 of the ATM pathway genes with pancreatic cancer risk. Am J Cancer Res 2020; 10:2128-2144. [PMID: 32775006 PMCID: PMC7407350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 06/21/2020] [Indexed: 06/11/2023] Open
Abstract
The ATM serine/threonine kinase (ATM) pathway plays important roles in pancreatic cancer (PanC) development and progression, but the roles of genetic variants of the genes in this pathway in the etiology of PanC are unknown. In the present study, we assessed associations between 31,499 single nucleotide polymorphisms (SNPs) in 198 ATM pathway-related genes and PanC risk using genotyping data from two previously published PanC genome-wide association studies (GWASs) of 15,423 subjects of European ancestry. In multivariable logistic regression analysis, we identified three novel independent SNPs to be significantly associated with PanC risk [PIK3C3 rs76692125 G>A: odds ratio (OR)=1.26, 95% confidence interval (CI)=1.12-1.43 and P=2.07×10-4, INSR rs11668724 G>A: OR=0.89, 95% CI=0.84-0.94 and P=4.21×10-5 and MAP3K4 rs13207108 C>T: OR=0.83, 95% CI=0.75-0.92, P=2.26×10-4]. The combined analysis of these three SNPs exhibited an increased PanC risk in a dose-response manner as the number of unfavorable genotypes increased (P trend<0.0001). The risk-associated rs76692125 A allele was correlated with decreased PIK3C3 mRNA expression levels, while the protective-associated rs11668724 A allele was correlated with increased INSR mRNA expression levels, but additional mechanistic studies of these SNPs are warranted. Once validated, these SNPs may serve as biomarkers for PanC risk in populations of European ancestry.
Collapse
Affiliation(s)
- Ling-Ling Zhao
- Cancer Center, The First Hospital of Jilin UniversityChangchun, China
- Duke Cancer Institute, Duke University Medical CenteDurham, NC, USA
- Department of Medicine, Duke University School of MedicineDurham, NC, USA
| | - Hong-Liang Liu
- Duke Cancer Institute, Duke University Medical CenteDurham, NC, USA
- Department of Medicine, Duke University School of MedicineDurham, NC, USA
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of MedicineDurham, NC, USA
| | - Kyle M Walsh
- Duke Cancer Institute, Duke University Medical CenteDurham, NC, USA
- Department of Neurosurgery, Duke University School of MedicineDurham, NC, USA
| | - Wei Li
- Cancer Center, The First Hospital of Jilin UniversityChangchun, China
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical CenteDurham, NC, USA
- Department of Medicine, Duke University School of MedicineDurham, NC, USA
- Department of Population Health Sciences, Duke University School of MedicineDurham, NC, USA
| |
Collapse
|
38
|
Abstract
BACKGROUND Pancreatic β-cells adapt to high metabolic demand by expanding their β-cell mass and/or enhancing insulin secretion to maintain glucose homeostasis. Type 2 diabetes (T2D) is typically characterized by β-cell decompensation. SCOPE OF THE REVIEW The current review focuses on summarizing the "omics" and "epi-omics" approaches that particularly focus on addressing the β-cell adaptation to insulin resistance and T2D. MAJOR CONCLUSIONS The molecular mechanisms underlying successful versus compromised β-cell adaptation to insulin resistance are not entirely understood. The last decade has seen an exponential increase in the use of "omics" and "epi-omics" approaches to dissect pathophysiology of metabolic diseases. One recent example is the emergence of m6A mRNA methylation as a new layer of regulation of gene expression with the potential to impact diverse physiological processes in metabolic cells.
Collapse
Affiliation(s)
- Dario F De Jesus
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA; Harvard Stem Cell Institute, Boston, USA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA; Harvard Stem Cell Institute, Boston, USA.
| |
Collapse
|
39
|
Her TK, Lagakos WS, Brown MR, LeBrasseur NK, Rakshit K, Matveyenko AV. Dietary carbohydrates modulate metabolic and β-cell adaptation to high-fat diet-induced obesity. Am J Physiol Endocrinol Metab 2020; 318:E856-E865. [PMID: 32315211 PMCID: PMC7311673 DOI: 10.1152/ajpendo.00539.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Obesity is associated with several chronic comorbidities, one of which is type 2 diabetes mellitus (T2DM). The pathogenesis of obesity and T2DM is influenced by alterations in diet macronutrient composition, which regulate energy expenditure, metabolic function, glucose homeostasis, and pancreatic islet cell biology. Recent studies suggest that increased intake of dietary carbohydrates plays a previously underappreciated role in the promotion of obesity and consequent metabolic dysfunction. Thus, in this study, we utilized mouse models to test the hypothesis that dietary carbohydrates modulate energetic, metabolic, and islet adaptions to high-fat diets. To address this, we exposed C57BL/6J mice to 12 wk of 3 eucaloric high-fat diets (>60% calories from fat) with varying total carbohydrate (1-20%) and sucrose (0-20%) content. Our results show that severe restriction of dietary carbohydrates characteristic of ketogenic diets reduces body fat accumulation, enhances energy expenditure, and reduces prevailing glycemia and insulin resistance compared with carbohydrate-rich, high-fat diets. Moreover, severe restriction of dietary carbohydrates also results in functional, morphological, and molecular changes in pancreatic islets highlighted by restricted capacity for β-cell mass expansion and alterations in insulin secretory response. These studies support the hypothesis that low-carbohydrate/high-fat diets provide antiobesogenic benefits and suggest further evaluation of the effects of these diets on β-cell biology in humans.
Collapse
Affiliation(s)
- Tracy K Her
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - William S Lagakos
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - Matthew R Brown
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - Nathan K LeBrasseur
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota
| | - Kuntol Rakshit
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - Aleksey V Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Rochester, Minnesota
- Department of Medicine, Division of Endocrinology, Metabolism, Diabetes, and Nutrition, Mayo Clinic School of Medicine, Rochester, Minnesota
| |
Collapse
|
40
|
Merry TL, Hedges CP, Masson SW, Laube B, Pöhlmann D, Wueest S, Walsh ME, Arnold M, Langhans W, Konrad D, Zarse K, Ristow M. Partial impairment of insulin receptor expression mimics fasting to prevent diet-induced fatty liver disease. Nat Commun 2020; 11:2080. [PMID: 32350271 PMCID: PMC7190665 DOI: 10.1038/s41467-020-15623-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 03/19/2020] [Indexed: 12/28/2022] Open
Abstract
Excessive insulin signaling through the insulin receptor (IR) may play a role in the pathogenesis of diet-induced metabolic disease, including obesity and type 2 diabetes. Here we investigate whether heterozygous impairment of insulin receptor (IR) expression limited to peripheral, i.e. non-CNS, tissues of adult mice impacts the development of high-fat diet-induced metabolic deterioration. While exhibiting some features of insulin resistance, PerIRKO+/− mice display a hepatic energy deficit accompanied by induction of energy-sensing AMPK, mitochondrial biogenesis, PPARα, unexpectedly leading to protection from, and reversal of hepatic lipid accumulation (steatosis hepatis, NAFLD). Consistently, and unlike in control mice, the PPARα activator fenofibrate fails to further affect hepatic lipid accumulation in PerIRKO+/− mice. Taken together, and opposing previously established diabetogenic features of insulin resistance, incomplete impairment of insulin signaling may mimic central aspects of calorie restriction to limit hepatic lipid accumulation during conditions of metabolic stress. Hyper-insulinemia associated with excess calorie intake may cause metabolic dysfunction. Here the authors report that mice with partially reduced insulin receptor expression in peripheral tissues are protected from and experience reversal of fatty liver disease.
Collapse
Affiliation(s)
- Troy L Merry
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland. .,Discipline of Nutrition, School of Medical Sciences, The University of Auckland, Auckland, New Zealand. .,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand.
| | - Chris P Hedges
- Discipline of Nutrition, School of Medical Sciences, The University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Stewart W Masson
- Discipline of Nutrition, School of Medical Sciences, The University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Beate Laube
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland
| | - Doris Pöhlmann
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland
| | - Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology and Children's Research Centre, University Children's Hospital, Zurich, Switzerland
| | - Michael E Walsh
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland
| | - Myrtha Arnold
- Physiology and Behavior Laboratory, Institute of Food and Nutrition, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Institute of Food and Nutrition, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology and Children's Research Centre, University Children's Hospital, Zurich, Switzerland
| | - Kim Zarse
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland
| | - Michael Ristow
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland.
| |
Collapse
|
41
|
Hu Z, Nizzero S, Goel S, Hinkle LE, Wu X, Li C, Ferrari M, Shen H. Molecular targeting of FATP4 transporter for oral delivery of therapeutic peptide. SCIENCE ADVANCES 2020; 6:eaba0145. [PMID: 32270048 PMCID: PMC7112756 DOI: 10.1126/sciadv.aba0145] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/09/2020] [Indexed: 06/11/2023]
Abstract
Low oral bioavailability of peptide drugs has limited their application to parenteral administration, which suffers from poor patient compliance. Here, we show that molecular targeting of the FATP4 transporter is an effective approach to specifically transport long-chain fatty acid (LCFA)-conjugated peptides across the enterocytic membrane and, thus, enables oral delivery of drug peptides. We packaged LCFA-conjugated exendin-4 (LCFA-Ex4) into liposomes and coated with chitosan nanoparticles to form an orally deliverable Ex4 (OraEx4). OraEx4 protected LCFA-Ex4 from damage by the gastric fluid and released LCFA-Ex4 in the intestinal cavity, where LCFA-Ex4 was transported across the enterocyte membrane by the FAPT4 transporter. OraEx4 had a high bioavailability of 24.8% with respect to subcutaneous injection and exhibited a substantial hypoglycemic effect in murine models of diabetes mellitus. Thus, molecular targeting of the FATP4 transporter enhances oral absorption of therapeutic peptides and provides a platform for oral peptide drug development.
Collapse
Affiliation(s)
- Zhenhua Hu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Sara Nizzero
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Shreya Goel
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Louis E. Hinkle
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Xiaoyan Wu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Pediatric Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chao Li
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
42
|
Rachdaoui N. Insulin: The Friend and the Foe in the Development of Type 2 Diabetes Mellitus. Int J Mol Sci 2020; 21:ijms21051770. [PMID: 32150819 PMCID: PMC7084909 DOI: 10.3390/ijms21051770] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/14/2022] Open
Abstract
Insulin, a hormone produced by pancreatic β-cells, has a primary function of maintaining glucose homeostasis. Deficiencies in β-cell insulin secretion result in the development of type 1 and type 2 diabetes, metabolic disorders characterized by high levels of blood glucose. Type 2 diabetes mellitus (T2DM) is characterized by the presence of peripheral insulin resistance in tissues such as skeletal muscle, adipose tissue and liver and develops when β-cells fail to compensate for the peripheral insulin resistance. Insulin resistance triggers a rise in insulin demand and leads to β-cell compensation by increasing both β-cell mass and insulin secretion and leads to the development of hyperinsulinemia. In a vicious cycle, hyperinsulinemia exacerbates the metabolic dysregulations that lead to β-cell failure and the development of T2DM. Insulin and IGF-1 signaling pathways play critical roles in maintaining the differentiated phenotype of β-cells. The autocrine actions of secreted insulin on β-cells is still controversial; work by us and others has shown positive and negative actions by insulin on β-cells. We discuss findings that support the concept of an autocrine action of secreted insulin on β-cells. The hypothesis of whether, during the development of T2DM, secreted insulin initially acts as a friend and contributes to β-cell compensation and then, at a later stage, becomes a foe and contributes to β-cell decompensation will be discussed.
Collapse
Affiliation(s)
- Nadia Rachdaoui
- Department of Animal Sciences, Room 108, Foran Hall, Rutgers, the State University of New Jersey, 59 Dudley Rd, New Brunswick, NJ 08901, USA
| |
Collapse
|
43
|
Shirakawa J, Tajima K, Okuyama T, Kyohara M, Togashi Y, De Jesus DF, Basile G, Kin T, Shapiro AMJ, Kulkarni RN, Terauchi Y. Luseogliflozin increases beta cell proliferation through humoral factors that activate an insulin receptor- and IGF-1 receptor-independent pathway. Diabetologia 2020; 63:577-587. [PMID: 31897526 PMCID: PMC7574158 DOI: 10.1007/s00125-019-05071-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Sodium-glucose cotransporter 2 (SGLT2) inhibitors, which prevent the renal reabsorption of glucose, decrease blood glucose levels in an insulin-independent manner. We previously reported creating a mouse model of systemic inhibition of target receptors for both insulin and IGF-1 by treating animals with OSI-906, a dual insulin/IGF-1 receptor inhibitor, for 7 days. The OSI-906-treated mice exhibited an increased beta cell mass, hepatic steatosis and adipose tissue atrophy, accompanied by hyperglycaemia and hyperinsulinaemia. In the present study, we investigated the effects of an SGLT2 inhibitor, luseogliflozin, on these changes in OSI-906-treated mice. METHODS We treated C57BL/6J male mice either with vehicle, luseogliflozin, OSI-906 or OSI-906 plus luseogliflozin for 7 days, and phenotyping was performed to determine beta cell mass and proliferation. Subsequently, we tested whether serum-derived factors have an effect on beta cell proliferation in genetically engineered beta cells, mouse islets or human islets. RESULTS SGLT2 inhibition with luseogliflozin significantly ameliorated hyperglycaemia, but not hyperinsulinaemia, in the OSI-906-treated mice. Liver steatosis and adipose tissue atrophy induced by OSI-906 were not altered by treatment with luseogliflozin. Beta cell mass and proliferation were further increased by SGLT2 inhibition with luseogliflozin in the OSI-906-treated mice. Luseogliflozin upregulated gene expression related to the forkhead box M1 (FoxM1)/polo-like kinase 1 (PLK1)/centromere protein A (CENP-A) pathway in the islets of OSI-906-treated mice. The increase in beta cell proliferation was recapitulated in a co-culture of Irs2 knockout and Insr/IR knockout (βIRKO) beta cells with serum from both luseogliflozin- and OSI-906-treated mice, but not after SGLT2 inhibition in beta cells. Circulating factors in both luseogliflozin- and OSI-906-treated mice promoted beta cell proliferation in both mouse islets and cadaveric human islets. CONCLUSIONS/INTERPRETATION These results suggest that luseogliflozin can increase beta cell proliferation through the activation of the FoxM1/PLK1/CENP-A pathway via humoral factors that act in an insulin/IGF-1 receptor-independent manner.
Collapse
Affiliation(s)
- Jun Shirakawa
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| | - Kazuki Tajima
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Tomoko Okuyama
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Mayu Kyohara
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Yu Togashi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Dario F De Jesus
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Giorgio Basile
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Tatsuya Kin
- Clinical Islet Laboratory and Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - A M James Shapiro
- Clinical Islet Laboratory and Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| |
Collapse
|
44
|
Maachi H, Fergusson G, Ethier M, Brill GN, Katz LS, Honig LB, Metukuri MR, Scott DK, Ghislain J, Poitout V. HB-EGF Signaling Is Required for Glucose-Induced Pancreatic β-Cell Proliferation in Rats. Diabetes 2020; 69:369-380. [PMID: 31882563 PMCID: PMC7034189 DOI: 10.2337/db19-0643] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 12/19/2019] [Indexed: 12/19/2022]
Abstract
The molecular mechanisms of β-cell compensation to metabolic stress are poorly understood. We previously observed that nutrient-induced β-cell proliferation in rats is dependent on epidermal growth factor receptor (EGFR) signaling. The aim of this study was to determine the role of the EGFR ligand heparin-binding EGF-like growth factor (HB-EGF) in the β-cell proliferative response to glucose, a β-cell mitogen and key regulator of β-cell mass in response to increased insulin demand. We show that exposure of isolated rat and human islets to HB-EGF stimulates β-cell proliferation. In rat islets, inhibition of EGFR or HB-EGF blocks the proliferative response not only to HB-EGF but also to glucose. Furthermore, knockdown of HB-EGF in rat islets blocks β-cell proliferation in response to glucose ex vivo and in vivo in transplanted glucose-infused rats. Mechanistically, we demonstrate that HB-EGF mRNA levels are increased in β-cells in response to glucose in a carbohydrate-response element-binding protein (ChREBP)-dependent manner. In addition, chromatin immunoprecipitation studies identified ChREBP binding sites in proximity to the HB-EGF gene. Finally, inhibition of Src family kinases, known to be involved in HB-EGF processing, abrogated glucose-induced β-cell proliferation. Our findings identify a novel glucose/HB-EGF/EGFR axis implicated in β-cell compensation to increased metabolic demand.
Collapse
Affiliation(s)
- Hasna Maachi
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, University of Montreal, Montreal, Quebec, Canada
| | - Grace Fergusson
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Melanie Ethier
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Gabriel N Brill
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Liora S Katz
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Lee B Honig
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Donald K Scott
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Julien Ghislain
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
45
|
Pradas-Juni M, Hansmeier NR, Link JC, Schmidt E, Larsen BD, Klemm P, Meola N, Topel H, Loureiro R, Dhaouadi I, Kiefer CA, Schwarzer R, Khani S, Oliverio M, Awazawa M, Frommolt P, Heeren J, Scheja L, Heine M, Dieterich C, Büning H, Yang L, Cao H, Jesus DFD, Kulkarni RN, Zevnik B, Tröder SE, Knippschild U, Edwards PA, Lee RG, Yamamoto M, Ulitsky I, Fernandez-Rebollo E, Vallim TQDA, Kornfeld JW. A MAFG-lncRNA axis links systemic nutrient abundance to hepatic glucose metabolism. Nat Commun 2020; 11:644. [PMID: 32005828 PMCID: PMC6994702 DOI: 10.1038/s41467-020-14323-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/27/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity and type 2 diabetes mellitus are global emergencies and long noncoding RNAs (lncRNAs) are regulatory transcripts with elusive functions in metabolism. Here we show that a high fraction of lncRNAs, but not protein-coding mRNAs, are repressed during diet-induced obesity (DIO) and refeeding, whilst nutrient deprivation induced lncRNAs in mouse liver. Similarly, lncRNAs are lost in diabetic humans. LncRNA promoter analyses, global cistrome and gain-of-function analyses confirm that increased MAFG signaling during DIO curbs lncRNA expression. Silencing Mafg in mouse hepatocytes and obese mice elicits a fasting-like gene expression profile, improves glucose metabolism, de-represses lncRNAs and impairs mammalian target of rapamycin (mTOR) activation. We find that obesity-repressed LincIRS2 is controlled by MAFG and observe that genetic and RNAi-mediated LincIRS2 loss causes elevated blood glucose, insulin resistance and aberrant glucose output in lean mice. Taken together, we identify a MAFG-lncRNA axis controlling hepatic glucose metabolism in health and metabolic disease. Despite widespread transcription of LncRNA in mammalian systems, their contribution to metabolic homeostasis at the cellular and tissue level remains elusive. Here Pradas-Juni et al. describe a transcription factor–LncRNA pathway that couples hepatocyte nutrient sensing to regulation of glucose metabolism in mice.
Collapse
Affiliation(s)
- Marta Pradas-Juni
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.,Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Nils R Hansmeier
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Jenny C Link
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA.,Department of Medicine, Division of Cardiology, UCLA, 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Elena Schmidt
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Bjørk Ditlev Larsen
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Paul Klemm
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Nicola Meola
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Hande Topel
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.,Izmir Biomedicine and Genome Center (IBG), Mithatpasa Ave. 58/5, 35340, Izmir, Turkey.,Department of Medical Biology and Genetics, Graduate School of Health Sciences, Dokuz Eylul University, Mithatpasa Ave. 1606, 35330, Izmir, Turkey
| | - Rute Loureiro
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.,Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Ines Dhaouadi
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Christoph A Kiefer
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Robin Schwarzer
- Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Sajjad Khani
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Matteo Oliverio
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Motoharu Awazawa
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, 162-8655, Japan
| | - Peter Frommolt
- Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, Martinistraße 52, 20246, Hamburg, Germany
| | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, Martinistraße 52, 20246, Hamburg, Germany
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, Martinistraße 52, 20246, Hamburg, Germany
| | - Christoph Dieterich
- Section of Bioinformatics and Systems Cardiology, Klaus Tschira Institute for Integrative Computational Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Hildegard Büning
- Institute of Experimental Hematology, Hanover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Ling Yang
- Cardiovascular Branch, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA.,Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Haiming Cao
- Cardiovascular Branch, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA
| | - Dario F De Jesus
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, 02215, MA, USA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, 02215, MA, USA
| | - Branko Zevnik
- CECAD in vivo Research Facility, Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Simon E Tröder
- CECAD in vivo Research Facility, Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, University Hospital Ulm, Albert-Einstein Allee 93, 89081, Ulm, Germany
| | - Peter A Edwards
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA.,Department of Medicine, Division of Cardiology, UCLA, 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | | | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku Medical Megabank Organization, Sendai, 980-8573, Japan
| | - Igor Ulitsky
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Eduardo Fernandez-Rebollo
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Thomas Q de Aguiar Vallim
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA. .,Department of Medicine, Division of Cardiology, UCLA, 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA.
| | - Jan-Wilhelm Kornfeld
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark. .,Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany. .,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany.
| |
Collapse
|
46
|
Abstract
Insulin secretion by the pancreatic β-cells is elicited in response to elevated extracellular glucose concentration. In addition to triggering insulin secretion, glucose-induced signal regulates β-cell proliferation and survival. However, the molecular mechanism underlying the effects of glucose on the β-cell functionality still remains unclear. Glucokinase, a hexokinase isozyme that catalyzes the phosphorylation of glucose, acts as the glucose sensor in the β-cells. To investigate the mechanisms of glucose signaling in the regulation of β-cell functions, we analyzed the role of glucokinase in insulin secretion, β-cell proliferation and β-cell apoptosis, using β-cell-specific glucokinase-haploinsufficient (Gck+/-) mice and allosteric glucokinase activators (GKAs). Glucokinase-mediated glucose metabolism (1) suppresses endoplasmic reticulum (ER) stress-induced β-cell apoptosis via inducing insulin receptor substrate-2 (IRS-2) expression and expression of ER stress-related molecules, (2) promotes adaptive β-cell proliferation through activation of the Forkhead Box M1 (FoxM1)/polo-like kinase-1 (PLK1)/centromere protein-A (CENP-A) pathway, (3) induces islet inflammation by promoting interaction of islet-derived S100 calcium-binding protein A8 (S100A8) with macrophages, (4) induces the expression of Fibulin-5 (Fbln5), an extracellular matrix protein to regulate β-cell functions, and (5) activates other unknown pathways. Glucagon-like peptide-1 (GLP-1) receptor agonists and dipeptidyl peptidase 4 (DPP-4) inhibitors have been found to possibly compensate for dysregulation of glucose metabolism in the β-cells. This review provides an update and overview of the recent advances in the study of β-cell pathophysiology and some therapeutic possibilities focusing on glucose-/glucokinase-mediated signaling.
Collapse
Affiliation(s)
- Jun Shirakawa
- Department of Endocrinology and Metabolism, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
47
|
Huo K, Li X, Hu W, Song X, Zhang D, Zhang X, Chen X, Yuan J, Zuo J, Wang X. RFRP-3, the Mammalian Ortholog of GnIH, Is a Novel Modulator Involved in Food Intake and Glucose Homeostasis. Front Endocrinol (Lausanne) 2020; 11:194. [PMID: 32328034 PMCID: PMC7160250 DOI: 10.3389/fendo.2020.00194] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 03/18/2020] [Indexed: 11/13/2022] Open
Abstract
RF amide-related peptide 3 (RFRP-3) is a reproductive inhibitor and an endogenous orexigenic neuropeptide that may be involved in energy homeostasis. In this study, we evaluated the effect of acute or chronic RFRP-3 treatment (administered via intraperitoneal injection) on the food intake, meal microstructure and weight of rats, as well as the mechanism through which RFRP-3 is involved in glucose metabolism in the pancreas and glucose disposal tissues of rat in vivo. Our results showed that the intraperitoneal administration of RFRP-3 to rats resulted in marked body mass increased, hyperphagia, hyperlipidemia, hyperglycemia, glucose intolerance, hypoinsulinism, hyperglucagon, and insulin resistance, as well as significant increases in the size of pancreatic islets and the inflammatory reaction. Thus, we strongly assert that RFRP-3 as a novel neuroendocrine regulator involved in blood glucose homeostasis.
Collapse
|
48
|
Oakie A, Zhou L, Rivers S, Cheung C, Li J, Wang R. Postnatal knockout of beta cell insulin receptor impaired insulin secretion in male mice exposed to high-fat diet stress. Mol Cell Endocrinol 2020; 499:110588. [PMID: 31541682 DOI: 10.1016/j.mce.2019.110588] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/28/2019] [Accepted: 09/16/2019] [Indexed: 01/19/2023]
Abstract
The presence of insulin receptor (IR) on insulin-secreting beta cells suggests an autocrine regulatory role for insulin in its own signalling. Congenital beta cell-specific IR knockout (βIRKO) mouse studies have demonstrated the development of age-dependent glucose intolerance. We investigated the role of beta cell IR signalling specifically during postnatal life following undisturbed prenatal pancreatic development and maturation. We utilized a tamoxifen-inducible mouse insulin 1 promoter (MIP) driven Cre recombinase IR knockout mouse model (MIP-βIRKO) to achieve partial knockout of IR in islets and determine the functional role of beta cell IR in adult mice fed a control normal diet (ND) or 60% high-fat diet (HFD). At 24 weeks of age, MIP-βIRKO ND mice maintained glucose tolerance, insulin release, and unchanged beta cell mass when compared to control ND mice. In contrast, 24-week-old MIP-βIRKO mice demonstrated significant glucose intolerance and lower insulin release after 18 weeks of HFD feeding. A reduction in beta cell soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein expression, phosphorylated AktS473 and P70S6K1T389, and glucose transporter 2 (GLUT2) expression were also identified in MIP-βIRKO HFD islets. Overall, the postnatal knockout of beta cell IR in HFD-fed mice resulted in decreased expression of beta cell glucose-sensing and exocytotic proteins and a reduction in intracellular signalling. These findings highlight that IR expression in the adult islet is required to maintain beta cell function under hyperglycemic stress.
Collapse
Affiliation(s)
- Amanda Oakie
- Children's Health Research Institute, University of Western Ontario, London, ON, N6C 2V5, Canada; Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON, N6C 2V5, Canada
| | - Liangyi Zhou
- Children's Health Research Institute, University of Western Ontario, London, ON, N6C 2V5, Canada; Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON, N6C 2V5, Canada
| | - Sydney Rivers
- Children's Health Research Institute, University of Western Ontario, London, ON, N6C 2V5, Canada; Department of Physiology and Pharmacology, and University of Western Ontario, London, ON, N6C 2V5, Canada
| | - Christy Cheung
- Children's Health Research Institute, University of Western Ontario, London, ON, N6C 2V5, Canada; Department of Physiology and Pharmacology, and University of Western Ontario, London, ON, N6C 2V5, Canada
| | - Jinming Li
- Children's Health Research Institute, University of Western Ontario, London, ON, N6C 2V5, Canada; Department of Physiology and Pharmacology, and University of Western Ontario, London, ON, N6C 2V5, Canada
| | - Rennian Wang
- Children's Health Research Institute, University of Western Ontario, London, ON, N6C 2V5, Canada; Department of Physiology and Pharmacology, and University of Western Ontario, London, ON, N6C 2V5, Canada; Department of Medicine, University of Western Ontario, London, ON, N6C 2V5, Canada.
| |
Collapse
|
49
|
Jang HB, Go MJ, Park SI, Lee HJ, Cho SB. Chronic heavy alcohol consumption influences the association between genetic variants of GCK or INSR and the development of diabetes in men: A 12-year follow-up study. Sci Rep 2019; 9:20029. [PMID: 31882596 PMCID: PMC6934767 DOI: 10.1038/s41598-019-56011-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022] Open
Abstract
Chronic heavy alcohol consumption is a risk factor for diabetes, which is characterized by impaired β-cell function and insulin resistance. We aimed to determine whether the longitudinal associations between genetic variants of glucokinase (GCK) and insulin receptor (INSR) and the risk of developing diabetes were influenced by chronic heavy alcohol consumption. Data were obtained from the Korean Genome and Epidemiology Study. To identify candidate variants, 1,520 subjects (726 non-drinkers and 794 heavy drinkers) were included in the baseline cross-sectional study. After excluding patients with diabetes at baseline and those with insufficient data on diabetes incidence, prospective analyses were conducted in 773 subjects (353 non-drinkers and 420 heavy drinkers). In the baseline cross-sectional study, one SNP (rs758989) in GCK and four SNPs (rs7245757, rs1035942, rs1035940, and rs2042901) in INSR were selected as candidate SNPs that interact with alcohol to affect prediabetes and diabetes. We identified that these GCK and INSR polymorphisms are affected by chronic heavy alcohol consumption and have an effect on the incidence of diabetes. The incidence of diabetes was increased in chronic heavy alcohol drinkers carrying the C allele of GCK compared with never-drinkers with the C allele (HR, 2.15; 95% CI 1.30-3.57), and was increased in chronic heavy alcohol drinkers who were not carrying the INSR haplotype (-/-) compared with never-drinkers carrying the AACT haplotype (HR, 1.98; 95% CI 1.24-3.18). Moreover, we observed that the aggravating effects on the late insulin secretion (I/G120 and I/G AUC 60-120) in individuals who were chronic heavy drinkers with C allele of GCK. In the INSR haplotype, chronic heavy drinkers not carrying AACT were associated with lower disposition index. These results potentially suggest that chronic heavy alcohol consumption induce β-cell dysfunction partially mediated by decreased GCK expression or decline of insulin sensitivity via inhibition of INSR, thereby contributing to the development of diabetes.
Collapse
Affiliation(s)
- Han Byul Jang
- Center for Biomedical Science, Korea National Institute of Health, Cheongju, Chungcheongbuk-do, Republic of Korea
| | - Min Jin Go
- Center for Genome Science, Korea National Institute of Health, Cheongju, Chungcheongbuk-do, Republic of Korea
| | - Sang Ick Park
- Center for Biomedical Science, Korea National Institute of Health, Cheongju, Chungcheongbuk-do, Republic of Korea
| | - Hye-Ja Lee
- Center for Biomedical Science, Korea National Institute of Health, Cheongju, Chungcheongbuk-do, Republic of Korea.
| | - Seong Beom Cho
- Center for Genome Science, Korea National Institute of Health, Cheongju, Chungcheongbuk-do, Republic of Korea.
| |
Collapse
|
50
|
Moullé VS, Parnet P. Effects of Nutrient Intake during Pregnancy and Lactation on the Endocrine Pancreas of the Offspring. Nutrients 2019; 11:nu11112708. [PMID: 31717308 PMCID: PMC6893668 DOI: 10.3390/nu11112708] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 12/26/2022] Open
Abstract
The pancreas has an essential role in the regulation of glucose homeostasis by secreting insulin, the only hormone with a blood glucose lowering effect in mammals. Several circulating molecules are able to positively or negatively influence insulin secretion. Among them, nutrients such as fatty acids or amino acids can directly act on specific receptors present on pancreatic beta cells. Dietary intake, especially excessive nutrient intake, is known to modify energy balance in adults, resulting in pancreatic dysfunction. However, gestation and lactation are critical periods for fetal development and pup growth and specific dietary nutrients are required for optimal growth. Feeding alterations during these periods will impact offspring development and increase the risk of developing metabolic disorders in adulthood, leading to metabolic programming. This review will focus on the influence of nutrient intake during gestation and lactation periods on pancreas development and function in offspring, highlighting the molecular mechanism of imprinting on this organ.
Collapse
|