1
|
Solinas G, Becattini B. An adipoincretin effect links adipostasis with insulin secretion. Trends Endocrinol Metab 2024; 35:466-477. [PMID: 38861922 DOI: 10.1016/j.tem.2023.10.009] [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: 07/14/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 06/13/2024]
Abstract
The current paradigm for the insulin system focuses on the phenomenon of glucose-stimulated insulin secretion and insulin action on blood glucose control. This historical glucose-centric perspective may have introduced a conceptual bias in our understanding of insulin regulation. A body of evidence demonstrating that in vivo variations in blood glucose and insulin secretion can be largely dissociated motivated us to reconsider the fundamental design of the insulin system as a control system for metabolic homeostasis. Here, we propose that a minimal glucose-centric model does not accurately describe the physiological behavior of the insulin system and propose a new paradigm focusing on the effects of incretins, arguing that under fasting conditions, insulin is regulated by an adipoincretin effect.
Collapse
Affiliation(s)
- Giovanni Solinas
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Barbara Becattini
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
2
|
Becattini B, Molinaro A, Henricsson M, Borén J, Solinas G. Adipocyte PI3K links adipostasis with baseline insulin secretion at fasting through an adipoincretin effect. Cell Rep 2024; 43:114132. [PMID: 38656871 DOI: 10.1016/j.celrep.2024.114132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/06/2024] [Accepted: 04/04/2024] [Indexed: 04/26/2024] Open
Abstract
Insulin-PI3K signaling controls insulin secretion. Understanding this feedback mechanism is crucial for comprehending how insulin functions. However, the role of adipocyte insulin-PI3K signaling in controlling insulin secretion in vivo remains unclear. Using adipocyte-specific PI3Kα knockout mice (PI3KαAdQ) and a panel of isoform-selective PI3K inhibitors, we show that PI3Kα and PI3Kβ activities are functionally redundant in adipocyte insulin signaling. PI3Kβ-selective inhibitors have no effect on adipocyte AKT phosphorylation in control mice but blunt it in adipocytes of PI3KαAdQ mice, demonstrating adipocyte-selective pharmacological PI3K inhibition in the latter. Acute adipocyte-selective PI3K inhibition increases serum free fatty acid (FFA) and potently induces insulin secretion. We name this phenomenon the adipoincretin effect. The adipoincretin effect operates in fasted mice with increasing FFA and decreasing glycemia, indicating that it is not primarily a control system for blood glucose. This feedback control system defines the rates of adipose tissue lipolysis and chiefly controls basal insulin secretion during fasting.
Collapse
Affiliation(s)
- Barbara Becattini
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Angela Molinaro
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Henricsson
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Giovanni Solinas
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
3
|
Chang H, Bennett AM, Cameron WD, Floro E, Au A, McFaul CM, Yip CM, Rocheleau JV. Targeting Apollo-NADP + to Image NADPH Generation in Pancreatic Beta-Cell Organelles. ACS Sens 2022; 7:3308-3317. [PMID: 36269889 PMCID: PMC9706804 DOI: 10.1021/acssensors.2c01174] [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] [Indexed: 01/31/2023]
Abstract
NADPH/NADP+ redox state supports numerous reactions related to cell growth and survival; yet the full impact is difficult to appreciate due to organelle compartmentalization of NADPH and NADP+. To study glucose-stimulated NADPH production in pancreatic beta-cell organelles, we targeted the Apollo-NADP+ sensor by first selecting the most pH-stable version of the single-color sensor. We subsequently targeted mTurquoise2-Apollo-NADP+ to various organelles and confirmed activity in the cytoplasm, mitochondrial matrix, nucleus, and peroxisome. Finally, we measured the glucose- and glutamine-stimulated NADPH responses by single- and dual-color imaging of the targeted sensors. Overall, we developed multiple organelle-targeted Apollo-NADP+ sensors to reveal the prominent role of beta-cell mitochondria in determining NADPH production in the cytoplasm, nucleus, and peroxisome.
Collapse
Affiliation(s)
- Huntley
H. Chang
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada,Toronto
General Hospital Research Institute, University
Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Alex M. Bennett
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada,Toronto
General Hospital Research Institute, University
Health Network, Toronto, Ontario M5G 2C4, Canada
| | - William D. Cameron
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada,Toronto
General Hospital Research Institute, University
Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Eric Floro
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada,Toronto
General Hospital Research Institute, University
Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Aaron Au
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Christopher M. McFaul
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Christopher M. Yip
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Jonathan V. Rocheleau
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada,Toronto
General Hospital Research Institute, University
Health Network, Toronto, Ontario M5G 2C4, Canada,Department
of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada,Banting
and Best Diabetes Centre, University of
Toronto, Toronto, Ontario M5G 2C4, Canada,
| |
Collapse
|
4
|
Liebmann M, Grupe K, Asuaje Pfeifer M, Rustenbeck I, Scherneck S. Differences in lipid metabolism in acquired versus preexisting glucose intolerance during gestation: role of free fatty acids and sphingosine-1-phosphate. Lipids Health Dis 2022; 21:99. [PMID: 36209101 PMCID: PMC9547403 DOI: 10.1186/s12944-022-01706-x] [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: 06/30/2022] [Accepted: 09/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The prevalence of gestational diabetes mellitus (GDM) is increasing worldwide. There is increasing evidence that GDM is a heterogeneous disease with different subtypes. An important question in this context is whether impaired glucose tolerance (IGT), which is a typical feature of the disease, may already be present before pregnancy and manifestation of the disease. The latter type resembles in its clinical manifestation prediabetes that has not yet manifested as type 2 diabetes (T2DM). Altered lipid metabolism plays a crucial role in the disorder's pathophysiology. The aim was to investigate the role of lipids which are relevant in diabetes-like phenotypes in these both models with different time of initial onset of IGT. METHODS Two rodent models reflecting different characteristics of human GDM were used to characterize changes in lipid metabolism occurring during gestation. Since the New Zealand obese (NZO)-mice already exhibit IGT before and during gestation, they served as a subtype model for GDM with preexisting IGT (preIGT) and were compared with C57BL/6 N mice with transient IGT acquired during gestation (aqIGT). While the latter model does not develop manifest diabetes even under metabolic stress conditions, the NZO mouse is prone to severe disease progression later in life. Metabolically healthy Naval Medical Research Institute (NMRI) mice served as controls. RESULTS In contrast to the aqIGT model, preIGT mice showed hyperlipidemia during gestation with elevated free fatty acids (FFA), triglycerides (TG), and increased atherogenic index. Interestingly, sphingomyelin (SM) concentrations in the liver decreased during gestation concomitantly with an increase in the sphingosine-1-phosphate (S1P) concentration in plasma. Further, preIGT mice showed impaired hepatic weight adjustment and alterations in hepatic FFA metabolism during gestation. This was accompanied by decreased expression of peroxisome proliferator-activated receptor alpha (PPARα) and lack of translocation of fatty acid translocase (FAT/CD36) to the hepatocellular plasma membrane. CONCLUSION The preIGT model showed impaired lipid metabolism both in plasma and liver, as well as features of insulin resistance consistent with increased S1P concentrations, and in these characteristics, the preIGT model differs from the common GDM subtype with aqIGT. Thus, concomitantly elevated plasma FFA and S1P concentrations, in addition to general shifts in sphingolipid fractions, could be an interesting signal that the metabolic disorder existed before gestation and that future pregnancies require more intensive monitoring to avoid complications. This graphical abstract was created with BioRender.com .
Collapse
Affiliation(s)
- Moritz Liebmann
- Institute of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, D-38106, Braunschweig, Germany
| | - Katharina Grupe
- Institute of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, D-38106, Braunschweig, Germany
| | - Melissa Asuaje Pfeifer
- Institute of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, D-38106, Braunschweig, Germany
| | - Ingo Rustenbeck
- Institute of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, D-38106, Braunschweig, Germany
| | - Stephan Scherneck
- Institute of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, D-38106, Braunschweig, Germany.
| |
Collapse
|
5
|
Naser W, Maymand S, Rivera LR, Connor T, Liongue C, Smith CM, Aston-Mourney K, McCulloch DR, McGee SL, Ward AC. Cytokine-inducible SH2 domain containing protein contributes to regulation of adiposity, food intake, and glucose metabolism. FASEB J 2022; 36:e22320. [PMID: 35470501 DOI: 10.1096/fj.202101882r] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/24/2022] [Accepted: 04/06/2022] [Indexed: 01/03/2023]
Abstract
The cytokine-inducible SH2 domain containing protein (CISH) is the founding member of the suppressor of cytokine signaling (SOCS) family of negative feedback regulators and has been shown to be a physiological regulator of signaling in immune cells. This study sought to investigate novel functions for CISH outside of the immune system. Mice deficient in CISH were generated and analyzed using a range of metabolic and other parameters, including in response to a high fat diet and leptin administration. CISH knockout mice possessed decreased body fat and showed resistance to diet-induced obesity. This was associated with reduced food intake, but unaltered energy expenditure and microbiota composition. CISH ablation resulted in reduced basal expression of the orexigenic Agrp gene in the arcuate nucleus (ARC) region of the brain. Cish was basally expressed in the ARC, with evidence of co-expression with the leptin receptor (Lepr) gene in Agrp-positive neurons. CISH-deficient mice also showed enhanced leptin responsiveness, although Cish expression was not itself modulated by leptin. CISH-deficient mice additionally exhibited improved insulin sensitivity on a high-fat diet, but not glucose tolerance despite reduced body weight. These data identify CISH as an important regulator of homeostasis through impacts on appetite control, mediated at least in part by negative regulation of the anorexigenic effects of leptin, and impacts on glucose metabolism.
Collapse
Affiliation(s)
- Wasan Naser
- School of Medicine, Deakin University, Geelong, Victoria, Australia.,College of Science, University of Baghdad, Baghdad, Iraq
| | - Saeed Maymand
- School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Leni R Rivera
- School of Medicine, Deakin University, Geelong, Victoria, Australia.,IMPACT, Deakin University, Geelong, Victoria, Australia
| | - Timothy Connor
- School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Clifford Liongue
- School of Medicine, Deakin University, Geelong, Victoria, Australia.,IMPACT, Deakin University, Geelong, Victoria, Australia
| | - Craig M Smith
- School of Medicine, Deakin University, Geelong, Victoria, Australia.,IMPACT, Deakin University, Geelong, Victoria, Australia
| | - Kathryn Aston-Mourney
- School of Medicine, Deakin University, Geelong, Victoria, Australia.,IMPACT, Deakin University, Geelong, Victoria, Australia
| | - Daniel R McCulloch
- School of Medicine, Deakin University, Geelong, Victoria, Australia.,IMPACT, Deakin University, Geelong, Victoria, Australia
| | - Sean L McGee
- School of Medicine, Deakin University, Geelong, Victoria, Australia.,IMPACT, Deakin University, Geelong, Victoria, Australia
| | - Alister C Ward
- School of Medicine, Deakin University, Geelong, Victoria, Australia.,IMPACT, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|
6
|
Bharath LP, Regan T, Conway R. Regulation of Immune Cell Function by Nicotinamide Nucleotide Transhydrogenase. Am J Physiol Cell Physiol 2022; 322:C666-C673. [PMID: 35138175 PMCID: PMC8977145 DOI: 10.1152/ajpcell.00607.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Redox homeostasis is elemental for the normal physiology of all cell types. Cells use multiple mechanisms to regulate the redox balance tightly. The onset and progression of many metabolic and aging-associated diseases occur due to the dysregulation of redox homeostasis. Thus, it is critical to identify and therapeutically target mechanisms that precipitate abnormalities in redox balance. Reactive oxygen species (ROS) produced within the immune cells regulate homeostasis, hyperimmune and hypoimmune cell responsiveness, apoptosis, immune response to pathogens, and tumor immunity. Immune cells have both cytosolic and organelle-specific redox regulatory systems to maintain appropriate levels of ROS. Nicotinamide nucleotide transhydrogenase (NNT) is an essential mitochondrial redox regulatory protein. Dysregulation of NNT function prevents immune cells from mounting an adequate immune response to pathogens, promotes a chronic inflammatory state associated with aging and metabolic diseases, and initiates conditions related to a dysregulated immune system such as autoimmunity. While many studies have reported on NNT in different cell types, including cancer cells, relatively few studies have explored NNT in immune cells. This review provides an overview of NNT and focuses on the current knowledge of NNT in the immune cells.
Collapse
Affiliation(s)
- Leena P Bharath
- Department of Nutrition and Public Health, Merrimack College, North Andover, Massachusetts, United States
| | - Thomas Regan
- Department of Nutrition and Public Health, Merrimack College, North Andover, Massachusetts, United States
| | - Rachel Conway
- Department of Nutrition and Public Health, Merrimack College, North Andover, Massachusetts, United States
| |
Collapse
|
7
|
Pohorec V, Križančić Bombek L, Skelin Klemen M, Dolenšek J, Stožer A. Glucose-Stimulated Calcium Dynamics in Beta Cells From Male C57BL/6J, C57BL/6N, and NMRI Mice: A Comparison of Activation, Activity, and Deactivation Properties in Tissue Slices. Front Endocrinol (Lausanne) 2022; 13:867663. [PMID: 35399951 PMCID: PMC8988149 DOI: 10.3389/fendo.2022.867663] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Although mice are a very instrumental model in islet beta cell research, possible phenotypic differences between strains and substrains are largely neglected in the scientific community. In this study, we show important phenotypic differences in beta cell responses to glucose between C57BL/6J, C57BL/6N, and NMRI mice, i.e., the three most commonly used strains. High-resolution multicellular confocal imaging of beta cells in acute pancreas tissue slices was used to measure and quantitatively compare the calcium dynamics in response to a wide range of glucose concentrations. Strain- and substrain-specific features were found in all three phases of beta cell responses to glucose: a shift in the dose-response curve characterizing the delay to activation and deactivation in response to stimulus onset and termination, respectively, and distinct concentration-encoding principles during the plateau phase in terms of frequency, duration, and active time changes with increasing glucose concentrations. Our results underline the significance of carefully choosing and reporting the strain to enable comparison and increase reproducibility, emphasize the importance of analyzing a number of different beta cell physiological parameters characterizing the response to glucose, and provide a valuable standard for future studies on beta cell calcium dynamics in health and disease in tissue slices.
Collapse
Affiliation(s)
- Viljem Pohorec
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | | | - Maša Skelin Klemen
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Jurij Dolenšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- *Correspondence: Andraž Stožer, ; Jurij Dolenšek,
| | - Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- *Correspondence: Andraž Stožer, ; Jurij Dolenšek,
| |
Collapse
|
8
|
Yang CH, Ann-Onda D, Lin X, Fynch S, Nadarajah S, Pappas EG, Liu X, Scott JW, Oakhill JS, Galic S, Shi Y, Moreno-Asso A, Smith C, Loudovaris T, Levinger I, Eizirik DL, Laybutt DR, Herzog H, Thomas HE, Loh K. Neuropeptide Y1 receptor antagonism protects β-cells and improves glycemic control in type 2 diabetes. Mol Metab 2021; 55:101413. [PMID: 34890851 PMCID: PMC8733231 DOI: 10.1016/j.molmet.2021.101413] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/12/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Loss of functional β-cell mass is a key factor contributing to poor glycemic control in advanced type 2 diabetes (T2D). We have previously reported that the inhibition of the neuropeptide Y1 receptor improves the islet transplantation outcome in type 1 diabetes (T1D). The aim of this study was to identify the pathophysiological role of the neuropeptide Y (NPY) system in human T2D and further evaluate the therapeutic potential of using the Y1 receptor antagonist BIBO3304 to improve β-cell function and survival in T2D. METHODS The gene expression of the NPY system in human islets from nondiabetic subjects and subjects with T2D was determined and correlated with the stimulation index. The glucose-lowering and β-cell-protective effects of BIBO3304, a selective orally bioavailable Y1 receptor antagonist, in high-fat diet (HFD)/multiple low-dose streptozotocin (STZ)-induced and genetically obese (db/db) T2D mouse models were assessed. RESULTS In this study, we identified a more than 2-fold increase in NPY1R and its ligand, NPY mRNA expression in human islets from subjects with T2D, which was significantly associated with reduced insulin secretion. Consistently, the pharmacological inhibition of Y1 receptors by BIBO3304 significantly protected β cells from dysfunction and death under multiple diabetogenic conditions in islets. In a preclinical study, we demonstrated that the inhibition of Y1 receptors by BIBO3304 led to reduced adiposity and enhanced insulin action in the skeletal muscle. Importantly, the Y1 receptor antagonist BIBO3304 treatment also improved β-cell function and preserved functional β-cell mass, thereby resulting in better glycemic control in both HFD/multiple low-dose STZ-induced and db/db T2D mice. CONCLUSIONS Our results revealed a novel causal link between increased islet NPY-Y1 receptor gene expression and β-cell dysfunction and failure in human T2D, contributing to the understanding of the pathophysiology of T2D. Furthermore, our results demonstrate that the inhibition of the Y1 receptor by BIBO3304 represents a potential β-cell-protective therapy for improving functional β-cell mass and glycemic control in T2D.
Collapse
Affiliation(s)
- Chieh-Hsin Yang
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia.
| | - Danise Ann-Onda
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia
| | - Xuzhu Lin
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia
| | - Stacey Fynch
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia
| | | | - Evan G Pappas
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia
| | - Xin Liu
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia
| | - John W Scott
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, 3000, Australia; The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
| | - Jonathan S Oakhill
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, 3000, Australia; Department of Medicine, University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Sandra Galic
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia; Department of Medicine, University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Yanchuan Shi
- Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, 2010, Australia; Faculty of Medicine, UNSW Australia, Sydney, 2052, Australia
| | - Alba Moreno-Asso
- Institute of Health and Sport (IHES), Victoria University, Footscray, VIC, Australia; Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, St Albans, VIC, Australia
| | - Cassandra Smith
- Institute of Health and Sport (IHES), Victoria University, Footscray, VIC, Australia; Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, St Albans, VIC, Australia
| | - Thomas Loudovaris
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia; Department of Medicine, University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Itamar Levinger
- Institute of Health and Sport (IHES), Victoria University, Footscray, VIC, Australia; Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, St Albans, VIC, Australia
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Universite Libre de Bruxelles (ULB), Brussels, Belgium; Indiana Biosciences Research Institute (IBRI), Indianapolis, IN, USA
| | - D Ross Laybutt
- Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, 2010, Australia; Faculty of Medicine, UNSW Australia, Sydney, 2052, Australia
| | - Herbert Herzog
- Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, 2010, Australia; Faculty of Medicine, UNSW Australia, Sydney, 2052, Australia
| | - Helen E Thomas
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia; Department of Medicine, University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Kim Loh
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia; Department of Medicine, University of Melbourne, Fitzroy, VIC, 3065, Australia.
| |
Collapse
|
9
|
Fan Y, Wolford BN, Lu H, Liang W, Sun J, Zhou W, Rom O, Mahajan A, Surakka I, Graham SE, Liu Z, Kim H, Ramdas S, Fritsche LG, Nielsen JB, Gabrielsen ME, Hveem K, Yang D, Song J, Garcia-Barrio MT, Zhang J, Liu W, Zhang K, Willer CJ, Chen YE. Type 2 diabetes sex-specific effects associated with E167K coding variant in TM6SF2. iScience 2021; 24:103196. [PMID: 34746691 PMCID: PMC8554487 DOI: 10.1016/j.isci.2021.103196] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/31/2021] [Accepted: 09/27/2021] [Indexed: 02/07/2023] Open
Abstract
The rs58542926C >T (E167K) variant of the transmembrane 6 superfamily member 2 gene (TM6SF2) is associated with increased risks for nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes (T2D). Nevertheless, the role of the TM6SF2 rs58542926 variant in glucose metabolism is poorly understood. We performed a sex-stratified analysis of the association between the rs58542926C >T variant and T2D in multiple cohorts. The E167K variant was significantly associated with T2D, especially in males. Using an E167K knockin (KI) mouse model, we found that male but not the female KI mice exhibited impaired glucose tolerance. As an ER membrane protein, TM6SF2 was found to interact with inositol-requiring enzyme 1 α (IRE1α), a primary ER stress sensor. The male Tm6sf2 KI mice exhibited impaired IRE1α signaling in the liver. In conclusion, the E167K variant of TM6SF2 is associated with glucose intolerance primarily in males, both in humans and mice.
Collapse
Affiliation(s)
- Yanbo Fan
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg 26, Rm 361S, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
- Department of Cancer Biology, University of Cincinnati College of Medicine, Vontz Center, 3125 Eden Avenue, Cincinnati, OH45267, USA
| | - Brooke N. Wolford
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI48109, USA
| | - Haocheng Lu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg 26, Rm 361S, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Wenying Liang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg 26, Rm 361S, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Jinjian Sun
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg 26, Rm 361S, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI48109, USA
| | - Oren Rom
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg 26, Rm 361S, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA71103, USA
| | - Anubha Mahajan
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ida Surakka
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg 26, Rm 361S, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Sarah E. Graham
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg 26, Rm 361S, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Zhipeng Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, USA
| | - Hyunbae Kim
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201, USA
| | - Shweta Ramdas
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Lars G. Fritsche
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Jonas B. Nielsen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg 26, Rm 361S, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Maiken Elvestad Gabrielsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Dongshan Yang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg 26, Rm 361S, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Jun Song
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg 26, Rm 361S, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Minerva T. Garcia-Barrio
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg 26, Rm 361S, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Jifeng Zhang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg 26, Rm 361S, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Wanqing Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201, USA
| | - Cristen J. Willer
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg 26, Rm 361S, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Y. Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg 26, Rm 361S, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| |
Collapse
|
10
|
Cartwright DM, Oakey LA, Fletcher RS, Doig CL, Heising S, Larner DP, Nasteska D, Berry CE, Heaselgrave SR, Ludwig C, Hodson DJ, Lavery GG, Garten A. Nicotinamide riboside has minimal impact on energy metabolism in mouse models of mild obesity. J Endocrinol 2021; 251:111-123. [PMID: 34370682 PMCID: PMC8494379 DOI: 10.1530/joe-21-0123] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/09/2021] [Indexed: 11/15/2022]
Abstract
Supplementation with precursors of NAD has been shown to prevent and reverse insulin resistance, mitochondrial dysfunction, and liver damage in mouse models of diet-induced obesity. We asked whether the beneficial effects of supplementation with the NAD precursor nicotinamide riboside (NR) are dependent on mouse strain. We compared the effects of NR supplementation on whole-body energy metabolism and mitochondrial function in mildly obese C57BL/6N and C57BL/6J mice, two commonly used strains to investigate metabolism. Male C57BL/6N and C57BL/6J mice were fed a high-fat diet (HFD) or standard chow with or without NR supplementation for 8 weeks. Body and organ weights, glucose tolerance, and metabolic parameters as well as mitochondrial O2 flux in liver and muscle fibers were assessed. We found that NR supplementation had no influence on body or organ weight, glucose metabolism or hepatic lipid accumulation, energy expenditure, or metabolic flexibility but increased mitochondrial respiration in soleus muscle in both mouse strains. Strain-dependent differences were detected for body and fat depot weight, fasting blood glucose, hepatic lipid accumulation, and energy expenditure. We conclude that, in mild obesity, NR supplementation does not alter metabolic phenotype in two commonly used laboratory mouse strains.
Collapse
Affiliation(s)
- David M Cartwright
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Lucy A Oakey
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Rachel S Fletcher
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Craig L Doig
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Silke Heising
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Dean P Larner
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Daniela Nasteska
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Caitlin E Berry
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Sam R Heaselgrave
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Christian Ludwig
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - David J Hodson
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Antje Garten
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Pediatric Research Center, Hospital for Child and Adolescent Medicine, Leipzig University, Leipzig, Germany
| |
Collapse
|
11
|
Gao R, Fu Q, Jiang HM, Shen M, Zhao RL, Qian Y, He YQ, Xu KF, Xu XY, Chen H, Zhang Q, Yang T. Temporal metabolic and transcriptomic characteristics crossing islets and liver reveal dynamic pathophysiology in diet-induced diabetes. iScience 2021; 24:102265. [PMID: 33817571 PMCID: PMC8008187 DOI: 10.1016/j.isci.2021.102265] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/30/2020] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
To investigate the molecular mechanisms underlying islet dysfunction and insulin resistance in diet-induced diabetes, we conducted temporal RNA sequencing of tissues responsible for insulin secretion (islets) and action (liver) every 4 weeks in mice on high-fat (HFD) or chow diet for 24 weeks, linking to longitudinal profile of metabolic characteristics. The diverse responses of α, β, and δ cells to glucose and palmitate indicated HFD-induced dynamic deterioration of islet function from dysregulation to failure. Insulin resistance developed with variable time course in different tissues. Weighted gene co-expression network analysis and Ingenuity Pathway Analysis implicated islets and liver jointly programmed β-cell compensatory adaption via cell proliferation at early phase and irreversible islet dysfunction by inappropriate immune response at later stage, and identified interconnected molecules including growth differentiation factor 15. Frequencies of T cell subpopulation showed an early decrement in Tregs followed by increases in Th1 and Th17 cells during progression to diabetes.
Collapse
Affiliation(s)
- Rui Gao
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China.,Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX37LE, UK
| | - Qi Fu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - He-Min Jiang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Min Shen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Rui-Ling Zhao
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yu Qian
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yun-Qiang He
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Kuan-Feng Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xin-Yu Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Heng Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Quan Zhang
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX37LE, UK
| | - Tao Yang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
12
|
Kunath A, Heiker JT, Kern M, Kosacka J, Flehmig G, Stumvoll M, Kovacs P, Blüher M, Klöting N. Nicotinamide Nucleotide Transhydrogenase (Nnt) is Related to Obesity in Mice. Horm Metab Res 2020; 52:877-881. [PMID: 32629517 DOI: 10.1055/a-1199-2257] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The C57BL/6J (B6J) mouse strain has been widely used as a control strain for the study of metabolic diseases and diet induced obesity (DIO). B6J mice carry a spontaneous deletion mutation in the nicotinamide nucleotide transhydrogenase (Nnt) gene eliminating exons 7-11, resulting in expression of a truncated form of Nnt, an enzyme that pumps protons across the inner mitochondrial membrane. It has been proposed that this mutation in B6J mice is associated with epigonadal fat mass and altered sensitivity to diet induced obesity. To define the role of Nnt in the development of diet induced obesity, we generated first backcross (BC1) hybrids of wild type Nnt C57BL/6NTac and mutated Nnt C57BL/6JRj [(C57BL/6NTac×C57BL/6JRj)F1×C57BL/6NTac]. Body weight gain and specific fat-pad depot mass were measured in BC1 hybrids under high fat diet conditions. Both sexes of BC1 hybrids indicate that mice with Nnt wild type allele are highly sensitive to DIO and exhibit higher relative fat mass. In summary, our data indicate that the Nnt mutation in mice is associated with sensitivity to DIO and fat mass.
Collapse
Affiliation(s)
- Anne Kunath
- IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Department of Medicine, Endocrinology and Diabetes, University of Leipzig, Leipzig, Germany
| | - John T Heiker
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| | - Matthias Kern
- Department of Medicine, Endocrinology and Diabetes, University of Leipzig, Leipzig, Germany
| | - Joanna Kosacka
- Department of Medicine, Endocrinology and Diabetes, University of Leipzig, Leipzig, Germany
| | - Gesine Flehmig
- Department of Medicine, Endocrinology and Diabetes, University of Leipzig, Leipzig, Germany
| | - Michael Stumvoll
- Department of Medicine, Endocrinology and Diabetes, University of Leipzig, Leipzig, Germany
| | - Peter Kovacs
- Interdisciplinary Center for Clinical Research, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Department of Medicine, Endocrinology and Diabetes, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| | - Nora Klöting
- IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Department of Medicine, Endocrinology and Diabetes, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| |
Collapse
|
13
|
Functions of Osteocalcin in Bone, Pancreas, Testis, and Muscle. Int J Mol Sci 2020; 21:ijms21207513. [PMID: 33053789 PMCID: PMC7589887 DOI: 10.3390/ijms21207513] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/05/2020] [Accepted: 10/10/2020] [Indexed: 12/12/2022] Open
Abstract
Osteocalcin (Ocn), which is specifically produced by osteoblasts, and is the most abundant non-collagenous protein in bone, was demonstrated to inhibit bone formation and function as a hormone, which regulates glucose metabolism in the pancreas, testosterone synthesis in the testis, and muscle mass, based on the phenotype of Ocn-/- mice by Karsenty's group. Recently, Ocn-/- mice were newly generated by two groups independently. Bone strength is determined by bone quantity and quality. The new Ocn-/- mice revealed that Ocn is not involved in the regulation of bone formation and bone quantity, but that Ocn regulates bone quality by aligning biological apatite (BAp) parallel to the collagen fibrils. Moreover, glucose metabolism, testosterone synthesis and spermatogenesis, and muscle mass were normal in the new Ocn-/- mice. Thus, the function of Ocn is the adjustment of growth orientation of BAp parallel to the collagen fibrils, which is important for bone strength to the loading direction of the long bone. However, Ocn does not play a role as a hormone in the pancreas, testis, and muscle. Clinically, serum Ocn is a marker for bone formation, and exercise increases bone formation and improves glucose metabolism, making a connection between Ocn and glucose metabolism.
Collapse
|
14
|
Yang CH, Mangiafico SP, Waibel M, Loudovaris T, Loh K, Thomas HE, Morahan G, Andrikopoulos S. E2f8 and Dlg2 genes have independent effects on impaired insulin secretion associated with hyperglycaemia. Diabetologia 2020; 63:1333-1348. [PMID: 32356104 DOI: 10.1007/s00125-020-05137-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/14/2020] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS Reduced insulin secretion results in hyperglycaemia and diabetes involving a complex aetiology that is yet to be fully elucidated. Genetic susceptibility is a key factor in beta cell dysfunction and hyperglycaemia but the responsible genes have not been defined. The Collaborative Cross (CC) is a recombinant inbred mouse panel with diverse genetic backgrounds allowing the identification of complex trait genes that are relevant to human diseases. The aim of this study was to identify and characterise genes associated with hyperglycaemia. METHODS Using an unbiased genome-wide association study, we examined random blood glucose and insulin sensitivity in 53 genetically unique mouse strains from the CC population. The influences of hyperglycaemia susceptibility quantitative trait loci (QTLs) were investigated by examining glucose tolerance, insulin secretion, pancreatic histology and gene expression in the susceptible mice. Expression of candidate genes and their association with insulin secretion were examined in human islets. Mechanisms underlying reduced insulin secretion were studied in MIN6 cells using RNA interference. RESULTS Wide variations in blood glucose levels and the related metabolic traits (insulin sensitivity and body weight) were observed in the CC population. We showed that elevated blood glucose in the CC strains was not due to insulin resistance nor obesity but resulted from reduced insulin secretion. This insulin secretory defect was demonstrated to be independent of abnormalities in islet morphology, beta cell mass and pancreatic insulin content. Gene mapping identified the E2f8 (p = 2.19 × 10-15) and Dlg2 loci (p = 3.83 × 10-8) on chromosome 7 to be significantly associated with hyperglycaemia susceptibility. Fine mapping the implicated regions using congenic mice demonstrated that these two loci have independent effects on insulin secretion in vivo. Significantly, our results revealed that increased E2F8 and DLG2 gene expression are correlated with enhanced insulin secretory function in human islets. Furthermore, loss-of-function studies in MIN6 cells demonstrated that E2f8 is involved in insulin secretion through an ATP-sensitive K+ channel-dependent pathway, which leads to a 30% reduction in Abcc8 expression. Similarly, knockdown of Dlg2 gene expression resulted in impaired insulin secretion in response to glucose and non-glucose stimuli. CONCLUSIONS/INTERPRETATION Collectively, these findings suggest that E2F transcription factor 8 (E2F8) and discs large homologue 2 (DLG2) regulate insulin secretion. The CC resource enables the identification of E2f8 and Dlg2 as novel genes associated with hyperglycaemia due to reduced insulin secretion in pancreatic beta cells. Taken together, our results provide better understanding of the molecular control of insulin secretion and further support the use of the CC resource to identify novel genes relevant to human diseases.
Collapse
Affiliation(s)
- Chieh-Hsin Yang
- Department of Medicine (Austin Health), Austin Hospital, University of Melbourne, Level 7, Lance Townsend Building, Studley Road, Heidelberg, VIC, 3084, Australia.
- St Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, VIC, 3065, Australia.
| | - Salvatore P Mangiafico
- Department of Medicine (Austin Health), Austin Hospital, University of Melbourne, Level 7, Lance Townsend Building, Studley Road, Heidelberg, VIC, 3084, Australia
| | - Michaela Waibel
- St Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, VIC, 3065, Australia
| | - Thomas Loudovaris
- St Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, VIC, 3065, Australia
| | - Kim Loh
- St Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, VIC, 3065, Australia
| | - Helen E Thomas
- St Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, VIC, 3065, Australia
| | - Grant Morahan
- Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
| | - Sofianos Andrikopoulos
- Department of Medicine (Austin Health), Austin Hospital, University of Melbourne, Level 7, Lance Townsend Building, Studley Road, Heidelberg, VIC, 3084, Australia.
| |
Collapse
|
15
|
Berdous D, Berney X, Sanchez-Archidona AR, Jan M, Roujeau C, Lopez-Mejia IC, Mynatt R, Thorens B. A genetic screen identifies Crat as a regulator of pancreatic beta-cell insulin secretion. Mol Metab 2020; 37:100993. [PMID: 32298772 PMCID: PMC7225740 DOI: 10.1016/j.molmet.2020.100993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 11/28/2022] Open
Abstract
Objectives Glucose-stimulated insulin secretion is a critical function in the regulation of glucose homeostasis, and its deregulation is associated with the development of type 2 diabetes. Here, we performed a genetic screen using islets isolated from the BXD panel of advanced recombinant inbred (RI) lines of mice to search for novel regulators of insulin production and secretion. Methods Pancreatic islets were isolated from 36 RI BXD lines and insulin secretion was measured following exposure to 2.8 or 16.7 mM glucose with or without exendin-4. Islets from the same RI lines were used for RNA extraction and transcript profiling. Quantitative trait loci (QTL) mapping was performed for each secretion condition and combined with transcriptome data to prioritize candidate regulatory genes within the identified QTL regions. Functional studies were performed by mRNA silencing or overexpression in MIN6B1 cells and by studying mice and islets with beta-cell-specific gene inactivation. Results Insulin secretion under the 16.7 mM glucose plus exendin-4 condition was mapped significantly to a chromosome 2 QTL. Within this QTL, RNA-Seq data prioritized Crat (carnitine O-acetyl transferase) as a strong candidate regulator of the insulin secretion trait. Silencing Crat expression in MIN6B1 cells reduced insulin content and insulin secretion by ∼30%. Conversely, Crat overexpression enhanced insulin content and secretion by ∼30%. When islets from mice with beta-cell-specific Crat inactivation were exposed to high glucose, they displayed a 30% reduction of insulin content as compared to control islets. We further showed that decreased Crat expression in both MIN6B1 cells and pancreatic islets reduced the oxygen consumption rate in a glucose concentration-dependent manner. Conclusions We identified Crat as a regulator of insulin secretion whose action is mediated by an effect on total cellular insulin content; this effect also depends on the genetic background of the RI mouse lines. These data also show that in the presence of the stimulatory conditions used the insulin secretion rate is directly related to the insulin content. A QTL analysis in BXD mice identifies Crat as a regulator of insulin secretion. Crat regulates insulin content in MIN6B1 cells and pancreatic islets. Crat regulates glucose oxidation in MIN6B1 cells and pancreatic islets. Crat links glucose metabolism to the control of beta-cell insulin content. Insulin content limits insulin secretion in response to high glucose and exendin-4 level.
Collapse
Affiliation(s)
- Dassine Berdous
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Xavier Berney
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Ana Rodriguez Sanchez-Archidona
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; Vital-IT, Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland.
| | - Maxime Jan
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Clara Roujeau
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Isabel C Lopez-Mejia
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Randall Mynatt
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
16
|
Ge MQ, Yeung SC, Mak JCW, Ip MSM. Differential metabolic and inflammatory responses to intermittent hypoxia in substrains of lean and obese C57BL/6 mice. Life Sci 2019; 238:116959. [PMID: 31628916 DOI: 10.1016/j.lfs.2019.116959] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 12/22/2022]
Abstract
AIMS This study was to investigate the degree of susceptibility to intermittent hypoxia (IH), a hallmark of obstructive sleep apnea (OSA), between the two mice inbred lines C57BL/6N (6N) and C57BL/6J (6J). MATERIALS AND METHODS Four-week old male mice of 6N and 6J substrains (n = 8) were randomized to standard diet (SD) group or high fat (HF) diet group. At the age of 13-week, all two groups of mice were subjected to either air or IH (IH30; thirty hypoxic events per hour) for one week. KEY FINDINGS All mice fed with HF diet exhibited obesity with more body weight and fat mass (percentage to body weight) gain. IH reduced serum LDL, HDL and total cholesterol levels in lean 6J mice. In obese mice, IH lowered obesity-induced serum total cholesterol level in 6J substrain but raised further in 6N substrain. Furthermore, IH caused elevation of serum FFA and MDA levels, and pro-inflammatory cytokines MCP-1 and IL-6 levels in subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) of lean 6J but not lean 6N mice. There was reduced number of adipocytes and elevation of macrophages in SAT and VAT of HF-induced obese mice of both substrains. IH led to increased number of adipocytes and macrophages in SAT of lean 6J mice. SIGNIFICANCE The genetic difference between 6N and 6J mice may have direct impact on metabolic and inflammatory responses after IH. Therefore, attention must be given for the selection of C57BL mice substrains in the experimental IH-exposed mouse model.
Collapse
Affiliation(s)
- Meng Qin Ge
- Departments of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Sze Chun Yeung
- Departments of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Judith Choi Wo Mak
- Departments of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China; Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China.
| | - Mary Sau Man Ip
- Departments of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China.
| |
Collapse
|
17
|
Alquier T, Poitout V. Considerations and guidelines for mouse metabolic phenotyping in diabetes research. Diabetologia 2018; 61:526-538. [PMID: 29143855 PMCID: PMC5805661 DOI: 10.1007/s00125-017-4495-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/12/2017] [Indexed: 02/07/2023]
Abstract
Mice are the most commonly used species in preclinical research on the pathophysiology of metabolic diseases. Although they are extremely useful for identifying pathways, mechanisms and genes regulating glucose and energy homeostasis, the specificities of the various mouse models and methodologies used to investigate a metabolic phenotype can have a profound impact on experimental results and their interpretation. This review aims to: (1) describe the most commonly used experimental tests to assess glucose and energy homeostasis in mice; (2) provide some guidelines regarding the design, analysis and interpretation of these tests, as well as for studies using genetic models; and (3) identify important caveats and confounding factors that must be taken into account in the interpretation of findings.
Collapse
Affiliation(s)
- Thierry Alquier
- Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue Saint-Denis, Office R08-418, Montreal, QC, H2X 0A9, Canada.
- Department of Medicine, Université de Montréal, Montreal, QC, Canada.
| | - Vincent Poitout
- Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue Saint-Denis, Office R08-418, Montreal, QC, H2X 0A9, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
18
|
Vozenilek AE, Vetkoetter M, Green JM, Shen X, Traylor JG, Klein RL, Orr AW, Woolard MD, Krzywanski DM. Absence of Nicotinamide Nucleotide Transhydrogenase in C57BL/6J Mice Exacerbates Experimental Atherosclerosis. J Vasc Res 2018; 55:98-110. [PMID: 29455203 DOI: 10.1159/000486337] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 12/14/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Mitochondrial reactive oxygen species (ROS) contribute to inflammation and vascular remodeling during atherosclerotic plaque formation. C57BL/6N (6N) and C57BL/6J (6J) mice display distinct mitochondrial redox balance due to the absence of nicotinamide nucleotide transhydrogenase (NNT) in 6J mice. We hypothesize that differential NNT expression between these animals alters plaque development. METHODS 6N and 6J mice were treated with AAV8-PCSK9 (adeno-associated virus serotype 8/proprotein convertase subtilisin/kexin type 9) virus leading to hypercholesterolemia, increased low-density lipoprotein, and atherosclerosis in mice fed a high-fat diet (HFD). Mice were co-treated with the mitochondria-targeted superoxide dismutase mimetic MitoTEMPO to assess the contribution of mitochondrial ROS to atherosclerosis. RESULTS Baseline and HFD-induced vascular superoxide is increased in 6J compared to 6N mice. MitoTEMPO diminished superoxide in both groups demonstrating differential production of mitochondrial ROS among these strains. PCSK9 treatment and HFD led to similar increases in plasma lipids in both 6N and 6J mice. However, 6J animals displayed significantly higher levels of plaque formation. MitoTEMPO reduced plasma lipids but did not affect plaque formation in 6N mice. In contrast, MitoTEMPO surprisingly increased plaque formation in 6J mice. CONCLUSION These data indicate that loss of NNT increases vascular ROS production and exacerbates atherosclerotic plaque development.
Collapse
Affiliation(s)
- Aimee E Vozenilek
- Department of Microbiology and Immunology, School of Medicine, Shreveport, Louisiana, USA.,Center for Cardiovascular Disease and Sciences, Louisiana State University Health Sciences Center - Shreveport, Shreveport, Louisiana, USA
| | - Matthew Vetkoetter
- Department of Cellular Biology and Anatomy, School of Medicine, Shreveport, Louisiana, USA.,Center for Cardiovascular Disease and Sciences, Louisiana State University Health Sciences Center - Shreveport, Shreveport, Louisiana, USA
| | - Jonette M Green
- Department of Pathology and Translational Pathobiology, School of Medicine, Shreveport, Louisiana, USA.,Center for Cardiovascular Disease and Sciences, Louisiana State University Health Sciences Center - Shreveport, Shreveport, Louisiana, USA
| | - Xinggui Shen
- Department of Pathology and Translational Pathobiology, School of Medicine, Shreveport, Louisiana, USA.,Center for Cardiovascular Disease and Sciences, Louisiana State University Health Sciences Center - Shreveport, Shreveport, Louisiana, USA
| | - James G Traylor
- Department of Pathology and Translational Pathobiology, School of Medicine, Shreveport, Louisiana, USA
| | - Ronald L Klein
- Department of Pharmacology, Toxicology and Neuroscience, School of Medicine, Shreveport, Louisiana, USA.,Center for Cardiovascular Disease and Sciences, Louisiana State University Health Sciences Center - Shreveport, Shreveport, Louisiana, USA
| | - A Wayne Orr
- Department of Cellular Biology and Anatomy, School of Medicine, Shreveport, Louisiana, USA.,Department of Pathology and Translational Pathobiology, School of Medicine, Shreveport, Louisiana, USA.,Center for Cardiovascular Disease and Sciences, Louisiana State University Health Sciences Center - Shreveport, Shreveport, Louisiana, USA
| | - Matthew D Woolard
- Department of Microbiology and Immunology, School of Medicine, Shreveport, Louisiana, USA.,Center for Cardiovascular Disease and Sciences, Louisiana State University Health Sciences Center - Shreveport, Shreveport, Louisiana, USA
| | - David M Krzywanski
- Department of Cellular Biology and Anatomy, School of Medicine, Shreveport, Louisiana, USA.,Center for Cardiovascular Disease and Sciences, Louisiana State University Health Sciences Center - Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
19
|
Bergeron V, Ghislain J, Vivot K, Tamarina N, Philipson LH, Fielitz J, Poitout V. Deletion of Protein Kinase D1 in Pancreatic β-Cells Impairs Insulin Secretion in High-Fat Diet-Fed Mice. Diabetes 2018; 67:71-77. [PMID: 29038309 PMCID: PMC5741145 DOI: 10.2337/db17-0982] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/09/2017] [Indexed: 12/29/2022]
Abstract
Ββ-Cell adaptation to insulin resistance is necessary to maintain glucose homeostasis in obesity. Failure of this mechanism is a hallmark of type 2 diabetes (T2D). Hence, factors controlling functional β-cell compensation are potentially important targets for the treatment of T2D. Protein kinase D1 (PKD1) integrates diverse signals in the β-cell and plays a critical role in the control of insulin secretion. However, the role of β-cell PKD1 in glucose homeostasis in vivo is essentially unknown. Using β-cell-specific, inducible PKD1 knockout mice (βPKD1KO), we examined the role of β-cell PKD1 under basal conditions and during high-fat feeding. βPKD1KO mice under a chow diet presented no significant difference in glucose tolerance or insulin secretion compared with mice expressing the Cre transgene alone; however, when compared with wild-type mice, both groups developed glucose intolerance. Under a high-fat diet, deletion of PKD1 in β-cells worsened hyperglycemia, hyperinsulinemia, and glucose intolerance. This was accompanied by impaired glucose-induced insulin secretion both in vivo in hyperglycemic clamps and ex vivo in isolated islets from high-fat diet-fed βPKD1KO mice without changes in islet mass. This study demonstrates an essential role for PKD1 in the β-cell adaptive secretory response to high-fat feeding in mice.
Collapse
Affiliation(s)
- Valérie Bergeron
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Julien Ghislain
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada
| | - Kevin Vivot
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada
| | | | | | - Jens Fielitz
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Vincent Poitout
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montréal, Quebec, Canada
| |
Collapse
|
20
|
Parilla JH, Willard JR, Barrow BM, Zraika S. A Mouse Model of Beta-Cell Dysfunction as Seen in Human Type 2 Diabetes. J Diabetes Res 2018; 2018:6106051. [PMID: 29854823 PMCID: PMC5952555 DOI: 10.1155/2018/6106051] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/30/2018] [Accepted: 04/08/2018] [Indexed: 12/28/2022] Open
Abstract
Loss of first-phase insulin release is an early pathogenic feature of type 2 diabetes (T2D). Various mouse models exist to study T2D; however, few recapitulate the early β-cell defects seen in humans. We sought to develop a nongenetic mouse model of T2D that exhibits reduced first-phase insulin secretion without a significant deficit in pancreatic insulin content. C57BL/6J mice were fed 10% or 60% fat diet for three weeks, followed by three consecutive, once-daily intraperitoneal injections of the β-cell toxin streptozotocin (STZ; 30, 50, or 75 mg/kg) or vehicle. Four weeks after injections, the first-phase insulin response to glucose was reduced in mice when high-fat diet was combined with 30, 50, or 75 mg/kg STZ. This was accompanied by diminished second-phase insulin release and elevated fed glucose levels. Further, body weight gain, pancreatic insulin content, and β-cell area were decreased in high fat-fed mice treated with 50 and 75 mg/kg STZ, but not 30 mg/kg STZ. Low fat-fed mice were relatively resistant to STZ, with the exception of reduced pancreatic insulin content and β-cell area. Together, these data demonstrate that in high fat-fed mice, three once-daily injections of 30 mg/kg STZ produces a model of β-cell failure without insulin deficiency that may be useful in studies investigating the etiology and progression of human T2D.
Collapse
Affiliation(s)
- Jacqueline H. Parilla
- Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Joshua R. Willard
- Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Breanne M. Barrow
- Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Sakeneh Zraika
- Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
21
|
Abstract
The pancreatic β-cell secretes insulin in response to elevated plasma glucose. This review applies an external bioenergetic critique to the central processes of glucose-stimulated insulin secretion, including glycolytic and mitochondrial metabolism, the cytosolic adenine nucleotide pool, and its interaction with plasma membrane ion channels. The control mechanisms responsible for the unique responsiveness of the cell to glucose availability are discussed from bioenergetic and metabolic control standpoints. The concept of coupling factor facilitation of secretion is critiqued, and an attempt is made to unravel the bioenergetic basis of the oscillatory mechanisms controlling secretion. The need to consider the physiological constraints operating in the intact cell is emphasized throughout. The aim is to provide a coherent pathway through an extensive, complex, and sometimes bewildering literature, particularly for those unfamiliar with the field.
Collapse
Affiliation(s)
- David G Nicholls
- Buck Institute for Research on Aging, Novato, California; and Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University Diabetes Centre, Malmo, Sweden
| |
Collapse
|
22
|
Hull RL, Willard JR, Struck MD, Barrow BM, Brar GS, Andrikopoulos S, Zraika S. High fat feeding unmasks variable insulin responses in male C57BL/6 mouse substrains. J Endocrinol 2017; 233:53-64. [PMID: 28138002 PMCID: PMC5358546 DOI: 10.1530/joe-16-0377] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 01/30/2017] [Indexed: 12/22/2022]
Abstract
Mouse models are widely used for elucidating mechanisms underlying type 2 diabetes. Genetic background profoundly affects metabolic phenotype; therefore, selecting the appropriate model is critical. Although variability in metabolic responses between mouse strains is now well recognized, it also occurs within C57BL/6 mice, of which several substrains exist. This within-strain variability is poorly understood and could emanate from genetic and/or environmental differences. To better define the within-strain variability, we performed the first comprehensive comparison of insulin secretion from C57BL/6 substrains 6J, 6JWehi, 6NJ, 6NHsd, 6NTac and 6NCrl. In vitro, glucose-stimulated insulin secretion correlated with Nnt mutation status, wherein responses were uniformly lower in islets from C57BL/6J vs C57BL/6N mice. In contrast, in vivo insulin responses after 18 weeks of low fat feeding showed no differences among any of the six substrains. When challenged with a high-fat diet for 18 weeks, C57BL/6J substrains responded with a similar increase in insulin release. However, variability was evident among C57BL/6N substrains. Strikingly, 6NJ mice showed no increase in insulin release after high fat feeding, contributing to the ensuing hyperglycemia. The variability in insulin responses among high-fat-fed C57BL/6N mice could not be explained by differences in insulin sensitivity, body weight, food intake or beta-cell area. Rather, as yet unidentified genetic and/or environmental factor(s) are likely contributors. Together, our findings emphasize that caution should be exercised in extrapolating data from in vitro studies to the in vivo situation and inform on selecting the appropriate C57BL/6 substrain for metabolic studies.
Collapse
Affiliation(s)
- Rebecca L Hull
- Veterans Affairs Puget Sound Health Care SystemSeattle, Washington, USA
- Division of MetabolismEndocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Joshua R Willard
- Veterans Affairs Puget Sound Health Care SystemSeattle, Washington, USA
| | - Matthias D Struck
- Division of MetabolismEndocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Breanne M Barrow
- Veterans Affairs Puget Sound Health Care SystemSeattle, Washington, USA
| | - Gurkirat S Brar
- Veterans Affairs Puget Sound Health Care SystemSeattle, Washington, USA
| | - Sofianos Andrikopoulos
- Department of MedicineUniversity of Melbourne, Austin Hospital, Heidelberg, Victoria, Australia
| | - Sakeneh Zraika
- Veterans Affairs Puget Sound Health Care SystemSeattle, Washington, USA
- Division of MetabolismEndocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
23
|
Leskov I, Neville A, Shen X, Pardue S, Kevil CG, Granger DN, Krzywanski DM. Nicotinamide nucleotide transhydrogenase activity impacts mitochondrial redox balance and the development of hypertension in mice. ACTA ACUST UNITED AC 2016; 11:110-121. [PMID: 28087333 DOI: 10.1016/j.jash.2016.12.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/02/2016] [Accepted: 12/06/2016] [Indexed: 02/07/2023]
Abstract
Oxidant stress contributes to the initiation and progression of hypertension (HTN) by enhancing endothelial dysfunction and/or causing perturbations in nitric oxide homeostasis. Differences in mitochondrial function may augment this process and provide insight into why age of onset and clinical outcomes differ among individuals from distinct ethnic groups. We have previously demonstrated that variation in normal mitochondrial function and oxidant production exists in endothelial cells from individuals of Caucasian and African-American ethnicity and that this variation contributes to endothelial dysfunction. To model these distinct mitochondrial redox phenotypes, we used C57Bl/6N (6N) and C57Bl/6J (6J) mice that also display unique mitochondrial functional properties due to the differential expression nicotinamide nucleotide transhydrogenase (NNT). We demonstrate that the absence of NNT in 6J cells led to distinct mitochondrial bioenergetic profiles and a pro-oxidative mitochondrial phenotype characterized by increased superoxide production and reduced glutathione peroxidase activity. Interestingly, we found that 6J animals have significantly higher systolic blood pressure compared to 6N animals, and this difference is exacerbated by angiotensin II treatment. The changes in pressure were accompanied by both mitochondrial and vascular dysfunction revealed by impaired respiratory control ratios and endothelial-dependent vessel dilation. All end points could be significantly ameliorated by treatment with the mitochondria-targeted superoxide dismutase mimetic MitoTEMPO demonstrating a critical role for the production of mitochondrial reactive oxygen species in the development of HTN in these animals. Taken together, these data indicate that the absence of NNT leads to variation in mitochondrial function and contributes to a unique mitochondrial redox phenotype that influences susceptibility to HTN by contributing to endothelial and vascular dysfunction.
Collapse
Affiliation(s)
- Igor Leskov
- Department of Cellular Biology and Anatomy, School of Medicine, Shreveport, LA, USA; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Amber Neville
- Department of Cellular Biology and Anatomy, School of Medicine, Shreveport, LA, USA; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Xinggui Shen
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA; Department of Pathology, School of Medicine, Shreveport, LA, USA
| | - Sibile Pardue
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA; Department of Pathology, School of Medicine, Shreveport, LA, USA
| | - Christopher G Kevil
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA; Department of Pathology, School of Medicine, Shreveport, LA, USA
| | - D Neil Granger
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA; Department of Molecular and Cellular Physiology, School of Medicine, Shreveport, LA, USA
| | - David M Krzywanski
- Department of Cellular Biology and Anatomy, School of Medicine, Shreveport, LA, USA; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA.
| |
Collapse
|
24
|
Attané C, Peyot ML, Lussier R, Zhang D, Joly E, Madiraju SRM, Prentki M. Differential Insulin Secretion of High-Fat Diet-Fed C57BL/6NN and C57BL/6NJ Mice: Implications of Mixed Genetic Background in Metabolic Studies. PLoS One 2016; 11:e0159165. [PMID: 27403868 PMCID: PMC4942110 DOI: 10.1371/journal.pone.0159165] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 06/28/2016] [Indexed: 01/02/2023] Open
Abstract
Many metabolic studies employ tissue-specific gene knockout mice, which requires breeding of floxed gene mice, available mostly on C57BL/6N (NN) genetic background, with cre or Flp recombinase-expressing mice, available on C57BL/6J (JJ) background, resulting in the generation of mixed C57BL/6NJ (NJ) genetic background mice. Recent awareness of many genetic differences between NN and JJ strains including the deletion of nicotinamide nucleotide transhydrogenase (nnt), necessitates examination of the consequence of mixed NJ background on glucose tolerance, beta cell function and other metabolic parameters. Male mice with NN and NJ genetic background were fed with normal or high fat diets (HFD) for 12 weeks and glucose and insulin homeostasis were studied. Genotype had no effect on body weight and food intake in mice fed normal or high fat diets. Insulinemia in the fed and fasted states and after a glucose challenge was lower in HFD-fed NJ mice, even though their glycemia and insulin sensitivity were similar to NN mice. NJ mice showed mild glucose intolerance. Moreover, glucose- but not KCl-stimulated insulin secretion in isolated islets was decreased in HFD-fed NJ vs NN mice without changes in insulin content and beta cell mass. Under normal diet, besides reduced fed insulinemia, NN and NJ mice presented similar metabolic parameters. However, HFD-fed NJ mice displayed lower fed and fasted insulinemia and glucose-induced insulin secretion in vivo and ex vivo, as compared to NN mice. These results strongly caution against using unmatched mixed genetic background C57BL/6 mice for comparisons, particularly under HFD conditions.
Collapse
Affiliation(s)
- Camille Attané
- Departments of Nutrition and Biochemistry, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montréal, QC, Canada
| | - Marie-Line Peyot
- Departments of Nutrition and Biochemistry, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montréal, QC, Canada
| | - Roxane Lussier
- Departments of Nutrition and Biochemistry, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montréal, QC, Canada
| | - Dongwei Zhang
- Departments of Nutrition and Biochemistry, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montréal, QC, Canada
| | - Erik Joly
- Departments of Nutrition and Biochemistry, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montréal, QC, Canada
| | - S. R. Murthy Madiraju
- Departments of Nutrition and Biochemistry, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montréal, QC, Canada
| | - Marc Prentki
- Departments of Nutrition and Biochemistry, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montréal, QC, Canada
- * E-mail:
| |
Collapse
|
25
|
A Chromosome 13 locus is associated with male-specific mortality in mice. Aging Clin Exp Res 2016; 28:59-67. [PMID: 25995165 DOI: 10.1007/s40520-015-0370-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 04/28/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND AND AIM Mortality is a highly complex trait influenced by a wide array of genetic factors. METHODS We examined a population of 1200 mice that were F2 generation offspring of a 4-way reciprocal cross between C57BL6/J and DBA2/J strains. Animals were sacrificed at age 200, 500, or 800 days and genotyped at 96 markers. The 800 days old cohort, which were the survivors of a much larger breeding group, were examined for enriched frequency of alleles that benefit survival and depletion of alleles that reduce survival. RESULTS Loci on Chr 13 in males and on Chr X in females were significantly distorted from Mendelian expectations, even after conservative correction for multiple testing. DBA2/J alleles between 35 and 80 Mb on Chr 13 were underrepresented in the age 800 male animals. D2 genotypes in this region were also associated with premature death during behavioral testing. Furthermore, confirmatory analysis showed BXD recombinant inbred strains carrying the D2 alleles in this region had shorter median survival. Exploration of available pathology data indicated that a syndrome involving dental malocclusions, pancreatic islet hypertrophy, and kidney lipidosis may have mediated the effects of DBA alleles on mortality specifically in male mice. The heterozygote advantage locus on the X Chr was not found to be associated with any pathology. CONCLUSIONS These results suggest a novel locus influencing survival in the B6/D2 genetic background, perhaps via a metabolic disorder that emerges by 200 days of age in male animals.
Collapse
|
26
|
Nolan CJ, Delghingaro-Augusto V. Reversibility of Defects in Proinsulin Processing and Islet β-Cell Failure in Obesity-Related Type 2 Diabetes. Diabetes 2016; 65:352-4. [PMID: 26798122 DOI: 10.2337/dbi15-0020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Christopher J Nolan
- Department of Endocrinology, Canberra Hospital, Woden, Australian Capital Territory, Australia Australian National University Medical School, Acton, Australian Capital Territory, Australia
| | | |
Collapse
|
27
|
Fontaine DA, Davis DB. Attention to Background Strain Is Essential for Metabolic Research: C57BL/6 and the International Knockout Mouse Consortium. Diabetes 2016; 65:25-33. [PMID: 26696638 PMCID: PMC4686949 DOI: 10.2337/db15-0982] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The International Knockout Mouse Consortium (IKMC) introduces its targeted constructs into C57BL/6N embryonic stem cells. However, breeding with a Cre-recombinase and/or Flp-recombinase mouse is required for the generation of a null allele with the IKMC cassette. Many recombinase strains are in the C57BL/6J background, resulting in knockout animals on a mixed strain background. This can lead to variability in metabolic data and the use of improper control groups. While C57BL/6N and C57BL/6J are derived from the same parental C57BL/6 strain, there are key genotypic and phenotypic differences between these substrains. Many researchers may not even be aware of these differences, as the shorthand C57BL/6 is often used to describe both substrains. We found that 58% of articles involving genetically modified mouse models did not completely address background strain. This review will describe these two substrains and highlight the importance of separate consideration in mouse model development. Our aim is to increase awareness of this issue in the diabetes research community and to provide practical strategies to enable researchers to avoid mixed strain animals when using IKMC knockout mice.
Collapse
Affiliation(s)
- Danielle A Fontaine
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI
| | - Dawn Belt Davis
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI William S. Middleton Memorial Veterans Hospital, Madison, WI
| |
Collapse
|
28
|
Jackson JB, Leung JH, Stout CD, Schurig-Briccio LA, Gennis RB. Review and Hypothesis. New insights into the reaction mechanism of transhydrogenase: Swivelling the dIII component may gate the proton channel. FEBS Lett 2015; 589:2027-33. [PMID: 26143375 DOI: 10.1016/j.febslet.2015.06.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/17/2015] [Accepted: 06/17/2015] [Indexed: 11/26/2022]
Abstract
The membrane protein transhydrogenase in animal mitochondria and bacteria couples reduction of NADP⁺ by NADH to proton translocation. Recent X-ray data on Thermus thermophilus transhydrogenase indicate a significant difference in the orientations of the two dIII components of the enzyme dimer (Leung et al., 2015). The character of the orientation change, and a review of information on the kinetics and thermodynamics of transhydrogenase, indicate that dIII swivelling might assist in the control of proton gating by the redox state of bound NADP⁺/NADPH during enzyme turnover.
Collapse
Affiliation(s)
- J Baz Jackson
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Josephine H Leung
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92307, USA
| | - Charles D Stout
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92307, USA
| | | | - Robert B Gennis
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
29
|
Picklo MJ, Thyfault JP. Vitamin E and vitamin C do not reduce insulin sensitivity but inhibit mitochondrial protein expression in exercising obese rats. Appl Physiol Nutr Metab 2014; 40:343-52. [PMID: 25761734 DOI: 10.1139/apnm-2014-0302] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Controversy exists as to whether supplementation with the antioxidants vitamin E and vitamin C blocks adaptation to exercise. Exercise is a first-line means to treat obesity and its complications. While diet-induced obesity alters mitochondrial function and induces insulin resistance (IR), no data exist as to whether supplementation with vitamin E and vitamin C modify responses to exercise in pre-existing obesity. We tested the hypothesis that dietary supplementation with vitamin E (0.4 g α-tocopherol acetate/kg) and vitamin C (0.5 g/kg) blocks exercise-induced improvements on IR and mitochondrial content in obese rats maintained on a high-fat (45% fat energy (en)) diet. Diet-induced obese, sedentary rats had a 2-fold higher homeostasis model assessment of insulin resistance and larger insulin area under the curve following glucose tolerances test than rats fed a low-fat (10% fat en) diet. Exercising (12 weeks at 5 times per week in a motorized wheel) of obese rats normalized IR indices, an effect not modified by vitamin E and vitamin C. Vitamin E and vitamin C supplementation with exercise elevated mtDNA content in adipose and skeletal muscle to a greater extent (20%) than exercise alone in a depot-specific manner. On the other hand, vitamin C and vitamin E decreased exercise-induced increases in mitochondrial protein content for complex I (40%) and nicotinamide nucleotide transhydrogenase (35%) in a muscle-dependent manner. These data indicate that vitamin E and vitamin C supplementation in obese rodents does not modify exercise-induced improvements in insulin sensitivity but that changes in mitochondrial biogenesis and mitochondrial protein expression may be modified by antioxidant supplementation.
Collapse
Affiliation(s)
- Matthew J Picklo
- USDA-ARS Grand Forks Human Nutrition Research Center, 2420 2nd Avenue North, Grand Forks, ND 58201, USA
| | | |
Collapse
|
30
|
GLUT4 defects in adipose tissue are early signs of metabolic alterations in Alms1GT/GT, a mouse model for obesity and insulin resistance. PLoS One 2014; 9:e109540. [PMID: 25299671 PMCID: PMC4192353 DOI: 10.1371/journal.pone.0109540] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 09/10/2014] [Indexed: 01/26/2023] Open
Abstract
Dysregulation of signaling pathways in adipose tissue leading to insulin resistance can contribute to the development of obesity-related metabolic disorders. Alström Syndrome, a recessive ciliopathy, caused by mutations in ALMS1, is characterized by progressive metabolic alterations such as childhood obesity, hyperinsulinemia, and type 2 diabetes. Here we investigated the role of Alms1 disruption in AT expansion and insulin responsiveness in a murine model for Alström Syndrome. A gene trap insertion in Alms1 on the insulin sensitive C57BL6/Ei genetic background leads to early hyperinsulinemia and a progressive increase in body weight. At 6 weeks of age, before the onset of the metabolic disease, the mutant mice had enlarged fat depots with hypertrophic adipocytes, but without signs of inflammation. Expression of lipogenic enzymes was increased. Pre-adipocytes isolated from mutant animals demonstrated normal adipogenic differentiation but gave rise to mature adipocytes with reduced insulin-stimulated glucose uptake. Assessment of whole body glucose homeostasis revealed glucose intolerance. Insulin stimulation resulted in proper AKT phosphorylation in adipose tissue. However, the total amount of glucose transporter 4 (SLC4A2) and its translocation to the plasma membrane were reduced in mutant adipose depots compared to wildtype littermates. Alterations in insulin stimulated trafficking of glucose transporter 4 are an early sign of metabolic dysfunction in Alström mutant mice, providing a possible explanation for the reduced glucose uptake and the compensatory hyperinsulinemia. The metabolic signaling deficits either reside downstream or are independent of AKT activation and suggest a role for ALMS1 in GLUT4 trafficking. Alström mutant mice represent an interesting model for the development of metabolic disease in which adipose tissue with a reduced glucose uptake can expand by de novo lipogenesis to an obese state.
Collapse
|
31
|
Fergusson G, Ethier M, Guévremont M, Chrétien C, Attané C, Joly E, Fioramonti X, Prentki M, Poitout V, Alquier T. Defective insulin secretory response to intravenous glucose in C57Bl/6J compared to C57Bl/6N mice. Mol Metab 2014; 3:848-54. [PMID: 25506550 PMCID: PMC4264561 DOI: 10.1016/j.molmet.2014.09.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 09/16/2014] [Accepted: 09/18/2014] [Indexed: 12/01/2022] Open
Abstract
Objective The C57Bl/6J (Bl/6J) mouse is the most widely used strain in metabolic research. This strain carries a mutation in nicotinamide nucleotide transhydrogenase (Nnt), a mitochondrial enzyme involved in NADPH production, which has been suggested to lead to glucose intolerance and beta-cell dysfunction. However, recent reports comparing Bl/6J to Bl/6N (carrying the wild-type Nnt allele) under normal diet have led to conflicting results using glucose tolerance tests. Thus, we assessed glucose-stimulated insulin secretion (GSIS), insulin sensitivity, clearance and central glucose-induced insulin secretion in Bl/6J and N mice using gold-standard methodologies. Methods GSIS was measured using complementary tests (oral and intravenous glucose tolerance tests) and hyperglycemic clamps. Whole-body insulin sensitivity was assessed using euglycemic-hyperinsulinemic clamps. Neurally-mediated insulin secretion was measured during central hyperglycemia. Results Bl/6J mice have impaired GSIS compared to Bl/6N when glucose is administered intravenously during both a tolerance test and hyperglycemic clamp, but not in response to oral glucose. First and second phases of GSIS are altered without changes in whole body insulin sensitivity, insulin clearance, beta-cell mass or central response to glucose, thereby demonstrating defective beta-cell function in Bl/6J mice. Conclusions The Bl/6J mouse strain displays impaired insulin secretion. These results have important implications for choosing the appropriate test to assess beta-cell function and background strain in genetically modified mouse models.
Collapse
Affiliation(s)
- Grace Fergusson
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada ; Rodent Metabolic Phenotyping Core of Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Canada
| | - Mélanie Ethier
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada ; Rodent Metabolic Phenotyping Core of Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Canada
| | - Mélanie Guévremont
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada ; Imaging and Cell Biology Core of Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Canada
| | - Chloé Chrétien
- Centre des Sciences du Goût et de l'Alimentation, Unité Mixte de Recherche CNRS, INRA, Université de Bourgogne, Dijon, France
| | - Camille Attané
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Erik Joly
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada ; Imaging and Cell Biology Core of Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Canada
| | - Xavier Fioramonti
- Centre des Sciences du Goût et de l'Alimentation, Unité Mixte de Recherche CNRS, INRA, Université de Bourgogne, Dijon, France
| | - Marc Prentki
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada ; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada ; Department of Nutrition, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada ; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada ; Department of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Thierry Alquier
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada ; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada ; Department of Pathology and Cell Biology, Université de Montréal, Montréal, QC H3T 1J4, Canada ; Department of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| |
Collapse
|
32
|
Global genomic and transcriptomic analysis of human pancreatic islets reveals novel genes influencing glucose metabolism. Proc Natl Acad Sci U S A 2014; 111:13924-9. [PMID: 25201977 DOI: 10.1073/pnas.1402665111] [Citation(s) in RCA: 347] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Genetic variation can modulate gene expression, and thereby phenotypic variation and susceptibility to complex diseases such as type 2 diabetes (T2D). Here we harnessed the potential of DNA and RNA sequencing in human pancreatic islets from 89 deceased donors to identify genes of potential importance in the pathogenesis of T2D. We present a catalog of genetic variants regulating gene expression (eQTL) and exon use (sQTL), including many long noncoding RNAs, which are enriched in known T2D-associated loci. Of 35 eQTL genes, whose expression differed between normoglycemic and hyperglycemic individuals, siRNA of tetraspanin 33 (TSPAN33), 5'-nucleotidase, ecto (NT5E), transmembrane emp24 protein transport domain containing 6 (TMED6), and p21 protein activated kinase 7 (PAK7) in INS1 cells resulted in reduced glucose-stimulated insulin secretion. In addition, we provide a genome-wide catalog of allelic expression imbalance, which is also enriched in known T2D-associated loci. Notably, allelic imbalance in paternally expressed gene 3 (PEG3) was associated with its promoter methylation and T2D status. Finally, RNA editing events were less common in islets than previously suggested in other tissues. Taken together, this study provides new insights into the complexity of gene regulation in human pancreatic islets and better understanding of how genetic variation can influence glucose metabolism.
Collapse
|
33
|
Chaudhury A. Similarity in Transcytosis of nNOSα in Enteric Nerve Terminals and Beta Cells of Pancreatic Islet. Front Med (Lausanne) 2014; 1:20. [PMID: 25705631 PMCID: PMC4335384 DOI: 10.3389/fmed.2014.00020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 07/15/2014] [Indexed: 02/06/2023] Open
Affiliation(s)
- Arun Chaudhury
- Division of Surgery, Brigham and Women's Hospital, Harvard Medical School and VA Boston HealthCare System , Boston, MA , USA
| |
Collapse
|
34
|
Mitochondrial genetic background modulates bioenergetics and susceptibility to acute cardiac volume overload. Biochem J 2013; 455:157-67. [PMID: 23924350 DOI: 10.1042/bj20130029] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Dysfunctional bioenergetics has emerged as a key feature in many chronic pathologies such as diabetes and cardiovascular disease. This has led to the mitochondrial paradigm in which it has been proposed that mtDNA sequence variation contributes to disease susceptibility. In the present study we show a novel animal model of mtDNA polymorphisms, the MNX (mitochondrial-nuclear exchange) mouse, in which the mtDNA from the C3H/HeN mouse has been inserted on to the C57/BL6 nuclear background and vice versa to test this concept. Our data show a major contribution of the C57/BL6 mtDNA to the susceptibility to the pathological stress of cardiac volume overload which is independent of the nuclear background. Mitochondria harbouring the C57/BL6J mtDNA generate more ROS (reactive oxygen species) and have a higher mitochondrial membrane potential relative to those with C3H/HeN mtDNA, independent of nuclear background. We propose this is the primary mechanism associated with increased bioenergetic dysfunction in response to volume overload. In summary, these studies support the 'mitochondrial paradigm' for the development of disease susceptibility, and show that the mtDNA modulates cellular bioenergetics, mitochondrial ROS generation and susceptibility to cardiac stress.
Collapse
|
35
|
Wong N, Morahan G, Stathopoulos M, Proietto J, Andrikopoulos S. A novel mechanism regulating insulin secretion involving Herpud1 in mice. Diabetologia 2013; 56:1569-76. [PMID: 23620059 DOI: 10.1007/s00125-013-2908-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 03/18/2013] [Indexed: 11/24/2022]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes results from beta cell dysfunction after prolonged physiological stress, which causes oversecretion of insulin. We recently found that insulin hypersecretion is mediated by at least two genes. Among mouse models of type 2 diabetes, the DBA/2 mouse strain is more susceptible to diabetes than is the C57BL/6J (B6J) strain. One distinctive feature of the DBA/2 mouse is that it hypersecretes insulin, independent of changes in insulin sensitivity; we identified Nnt as a gene responsible for this trait. METHODS To identify the other gene(s) affecting insulin hypersecretion, we tested a panel of recombinant inbred BXD strains, which have different combinations of B6 and DBA/2 alleles. RESULTS We found that 25% of the BXD strains hypersecreted insulin in response to glucose. Microarray profiling of islets from high- and low-secretor strains showed that at least four genes were differentially expressed. One gene was consistently underexpressed in islets from both DBA/2 and the high-secretor BXD strains. This gene (Herpud1 or Herp) encodes the 54 kDa endoplasmic reticulum stress-inducible protein (HERP) that resides in the integral endoplasmic reticulum membrane. To test directly whether Herpud1 can interact with Nnt, Herpud1 was either knocked down or overexpressed in MIN6 cells. These results showed that when Herpud1 was suppressed, Nnt expression was reduced, while overexpression of Herpud1 led to increased Nnt expression. Furthermore, Herpud1 suppression resulted in significantly decreased glucose-stimulated insulin secretion in the DBA/2 islets but not B6J islets. CONCLUSIONS/INTERPRETATION We conclude that Herpud1 regulates insulin secretion via control of Nnt expression.
Collapse
Affiliation(s)
- N Wong
- Department of Medicine (Austin Health), Austin Hospital, University of Melbourne, Heidelberg Heights, Melbourne, Victoria, 3084, Australia.
| | | | | | | | | |
Collapse
|
36
|
Heiker JT, Kern M, Kosacka J, Flehmig G, Stumvoll M, Shang E, Lohmann T, Dreßler M, Kovacs P, Blüher M, Klöting N. Nicotinamide nucleotide transhydrogenase mRNA expression is related to human obesity. Obesity (Silver Spring) 2013; 21:529-34. [PMID: 23592659 DOI: 10.1002/oby.20095] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 08/20/2012] [Indexed: 12/26/2022]
Abstract
OBJECTIVE A spontaneous deletion in the nicotinamide nucleotide transhydrogenase (Nnt) gene eliminating exons 7-11 in C57BL/6J (B6J) mice is associated with reduced glucose-stimulated insulin secretion in vitro, impaired glucose tolerance, higher epigonadal fat mass, and altered susceptibility to diet induced obesity of male B6J mice was proposed. A potential implication for NNT in human adipose tissue distribution has not been investigated so far. DESIGN AND METHODS Therefore, NNT mRNA expression in paired human samples of visceral (vis) and subcutaneous (sc) adipose tissue from 221 subjects with a wide range of body mass index (BMI), insulin sensitivity, and glucose tolerance was analyzed. RESULTS NNT mRNA expression is significantly higher in visceral fat of obese patients and correlates with body weight, BMI, % body fat, visceral and sc fat area, waist and hip circumference, and fasting plasma insulin (FPI). Multivariate linear regression analysis revealed visceral NNT expression as age and gender independent predictor of BMI, waist circumference, visceral fat area, and % body fat, but not FPI and 2 h OGTT glucose. CONCLUSION In conclusion, a functional relevance of NNT in the development of human obesity and visceral fat distribution was suggested here.
Collapse
Affiliation(s)
- John T Heiker
- Department of Medicine, Endocrinology and Diabetes, Universität Leipzig, Leipzig, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Carreras A, Kayali F, Zhang J, Hirotsu C, Wang Y, Gozal D. Metabolic effects of intermittent hypoxia in mice: steady versus high-frequency applied hypoxia daily during the rest period. Am J Physiol Regul Integr Comp Physiol 2012; 303:R700-9. [PMID: 22895743 DOI: 10.1152/ajpregu.00258.2012] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Intermittent hypoxia (IH) is a frequent occurrence in sleep and respiratory disorders. Both human and murine studies show that IH may be implicated in metabolic dysfunction. Although the effects of nocturnal low-frequency intermittent hypoxia (IH(L)) have not been extensively examined, it would appear that IH(L) and high-frequency intermittent hypoxia (IH(H)) may elicit distinct metabolic adaptations. To this effect, C57BL/6J mice were randomly assigned to IH(H) (cycles of 90 s 6.4% O(2) and 90 s 21% O(2) during daylight), IH(L) (8% O(2) during daylight hours), or control (CTL) for 5 wk. At the end of exposures, some of the mice were subjected to a glucose tolerance test (GTT; after intraperitoneal injection of 2 mg glucose/g body wt), and others were subjected to an insulin tolerance test (ITT; 0.25 units Humulin/kg body wt), with plasma leptin and insulin levels being measured in fasting conditions. Skeletal muscles were harvested for GLUT4 and proliferator-activated receptor gamma coactivator 1-α (PGC1-α) expression. Both IH(H) and IH(L) displayed reduced body weight increases compared with CTL. CTL mice had higher basal glycemic levels, but GTT kinetics revealed marked differences between IH(L) and IH(H), with IH(L) manifesting the lowest insulin sensitivity compared with either IH(H) or CTL, and such findings were further confirmed by ITT. No differences emerged in PGC1-α expression across the three experimental groups. However, while cytosolic GLUT4 protein expression remained similar in IH(L), IH(H), and CTL, significant decreases in GLUT4 membrane fraction occurred in hypoxia and were most pronounced in IH(L)-exposed mice. Thus IH(H) and IH(L) elicit differential glucose homeostatic responses despite similar cumulative hypoxic profiles.
Collapse
Affiliation(s)
- Alba Carreras
- Dept. of Pediatrics, Univ. of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | |
Collapse
|
38
|
Alonso LC, Watanabe Y, Stefanovski D, Lee EJ, Singamsetty S, Romano LC, Zou B, Garcia-Ocaña A, Bergman RN, O'Donnell CP. Simultaneous measurement of insulin sensitivity, insulin secretion, and the disposition index in conscious unhandled mice. Obesity (Silver Spring) 2012; 20:1403-12. [PMID: 22331130 PMCID: PMC3378770 DOI: 10.1038/oby.2012.36] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Of the parameters that determine glucose disposal and progression to diabetes in humans: first-phase insulin secretion, glucose effectiveness (Sg), insulin sensitivity (Si), and the disposition index (DI), only Si can be reliably measured in conscious mice. To determine the importance of the other parameters in murine glucose homeostasis in lean and obese states, we developed the frequently sampled intravenous glucose tolerance test (FSIVGTT) for use in unhandled mice. We validated the conscious FSIVGTT against the euglycemic clamp for measuring Si in lean and obese mice. Insulin-resistant mice had increased first-phase insulin secretion, decreased Sg, and a reduced DI, qualitatively similar to humans. Intriguingly, although insulin secretion explained most of the variation in glucose disposal in lean mice, Sg and the DI more strongly predicted glucose disposal in obese mice. DI curves identified individual diet-induced obese (DIO) mice as having compensated or decompensated insulin secretion. Conscious FSIVGTT opens the door to apply mouse genetics to the determinants of in vivo insulin secretion, Sg, and DI, and further validates the mouse as a model of metabolic disease.
Collapse
Affiliation(s)
- Laura C Alonso
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Jackson JB. A review of the binding-change mechanism for proton-translocating transhydrogenase. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2012; 1817:1839-46. [PMID: 22538293 DOI: 10.1016/j.bbabio.2012.04.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 04/04/2012] [Accepted: 04/10/2012] [Indexed: 11/17/2022]
Abstract
Proton-translocating transhydrogenase is found in the inner membranes of animal mitochondria, and in the cytoplasmic membranes of many bacteria. It catalyses hydride transfer from NADH to NADP(+) coupled to inward proton translocation. Evidence is reviewed suggesting the enzyme operates by a "binding-change" mechanism. Experiments with Escherichia coli transhydrogenase indicate the enzyme is driven between "open" and "occluded" states by protonation and deprotonation reactions associated with proton translocation. In the open states NADP(+)/NADPH can rapidly associate with, or dissociate from, the enzyme, and hydride transfer is prevented. In the occluded states bound NADP(+)/NADPH cannot dissociate, and hydride transfer is allowed. Crystal structures of a complex of the nucleotide-binding components of Rhodospirillum rubrum transhydrogenase show how hydride transfer is enabled and disabled at appropriate steps in catalysis, and how release of NADP(+)/NADPH is restricted in the occluded state. Thermodynamic and kinetic studies indicate that the equilibrium constant for hydride transfer on the enzyme is elevated as a consequence of the tight binding of NADPH relative to NADP(+). The protonation site in the translocation pathway must face the outside if NADP(+) is bound, the inside if NADPH is bound. Chemical shift changes detected by NMR may show where alterations in protein conformation resulting from NADP(+) reduction are initiated. This article is part of a Special Issue entitled: 17th European Bioenergetics Conference (EBEC 2012).
Collapse
|
40
|
Potter DA, Yee D, Guo Z, Rodriguez M. Should diabetic women with breast cancer have their own intervention studies? Endocr Relat Cancer 2012; 19:C13-7. [PMID: 22180498 PMCID: PMC3680369 DOI: 10.1530/erc-11-0309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This commentary on 'Calorie restriction and rapamycin inhibit MMTV-Wnt-1 mammary tumor growth in a mouse model of postmenopausal obesity' by Nogueira et al., published in this issue of Endocrine-Related Cancer, addresses the challenges of translating diet, exercise, and pharmacologic trials in diabetic mouse mammary tumor models to human studies. We propose that trials specifically designed to test such interventions in diabetic women with breast cancer would be valuable and informative.
Collapse
Affiliation(s)
- David A Potter
- Division of Hematology, Oncology and Transplantation, Masonic Cancer Center, University of Minnesota, 420 Delaware Street SouthEast, MMC 480, Minneapolis, Minnesota 55455-0392, USA.
| | | | | | | |
Collapse
|
41
|
Wong N, Fam BC, Cempako GR, Steinberg GR, Walder K, Kay TW, Proietto J, Andrikopoulos S. Deficiency in interferon-gamma results in reduced body weight and better glucose tolerance in mice. Endocrinology 2011; 152:3690-9. [PMID: 21791564 DOI: 10.1210/en.2011-0288] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Obesity is a chronic low-grade inflammatory disease caused by increased energy intake and reduced energy expenditure. Studies using animal models with deletion of inflammatory cytokines have produced conflicting results with some showing increased weight gain and others showing no effect or even reduced body weights. Clearly, more work is necessary to understand the role of cytokines on body weight control. The aim of this study was to determine the effect of interferon-γ deletion (IFNγ(-/-)) on body weight regulation and glucose metabolism. Male IFNγ(-/-) and wild-type C57BL/6 mice were fed a low-fat chow diet, and body weight, food intake, and energy expenditure were monitored over 20 wk. At the end of the study, ip glucose tolerance test, insulin tolerance test, basal glucose turnover, and hyperinsulinemic/euglycemic clamps were performed. Expression levels of arcuate nucleus neuropeptide Y, Agouti-related peptide, and proopiomelanocortin mRNA as well as circulating leptin levels were also determined. IFNγ(-/-) mice had improved glucose tolerance with reduced rate of glucose appearance and increased insulin sensitivity due to greater suppression of endogenous glucose output, which was associated with decreased hepatic glucose-6-phosphatase activity. In addition, we also observed reduced body weight associated with decreased food intake and increased physical activity. Neuropeptide Y and Agouti-related peptide mRNA expression was reduced, whereas proopiomelanocortin mRNA expression was increased, as were plasma leptin levels. Global deletion of IFNγ in mice resulted in reduced body weight associated with negative energy balance, improved glucose tolerance, and hepatic insulin sensitivity. Our findings demonstrate that IFNγ plays a critical role in the regulation of body weight and glucose metabolism.
Collapse
Affiliation(s)
- Nicole Wong
- University of Melbourne, Department of Medicine (Austin Health and Northern Health), Heidelberg Repatriation Hospital, 300 Waterdale Road, Heidelberg Heights, Victoria 3081, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Albracht SPJ, Meijer AJ, Rydström J. Mammalian NADH:ubiquinone oxidoreductase (Complex I) and nicotinamide nucleotide transhydrogenase (Nnt) together regulate the mitochondrial production of H₂O₂--implications for their role in disease, especially cancer. J Bioenerg Biomembr 2011; 43:541-64. [PMID: 21882037 DOI: 10.1007/s10863-011-9381-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 08/03/2011] [Indexed: 12/20/2022]
Abstract
Mammalian NADH:ubiquinone oxidoreductase (Complex I) in the mitochondrial inner membrane catalyzes the oxidation of NADH in the matrix. Excess NADH reduces nine of the ten prosthetic groups of the enzyme in bovine-heart submitochondrial particles with a rate of at least 3,300 s⁻¹. This results in an overall NADH→O₂ rate of ca. 150 s⁻¹. It has long been known that the bovine enzyme also has a specific reaction site for NADPH. At neutral pH excess NADPH reduces only three to four of the prosthetic groups in Complex I with a rate of 40 s⁻¹ at 22 °C. The reducing equivalents remain essentially locked in the enzyme because the overall NADPH→O₂ rate (1.4 s⁻¹) is negligible. The physiological significance of the reaction with NADPH is still unclear. A number of recent developments has revived our thinking about this enigma. We hypothesize that Complex I and the Δp-driven nicotinamide nucleotide transhydrogenase (Nnt) co-operate in an energy-dependent attenuation of the hydrogen-peroxide generation by Complex I. This co-operation is thought to be mediated by the NADPH/NADP⁺ ratio in the vicinity of the NADPH site of Complex I. It is proposed that the specific H₂O₂ production by Complex I, and the attenuation of it, is of importance for apoptosis, autophagy and the survival mechanism of a number of cancers. Verification of this hypothesis may contribute to a better understanding of the regulation of these processes.
Collapse
Affiliation(s)
- Simon P J Albracht
- Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, NL-1098 XH, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
43
|
Mangiafico SP, Lim SH, Neoh S, Massinet H, Joannides CN, Proietto J, Andrikopoulos S, Fam BC. A primary defect in glucose production alone cannot induce glucose intolerance without defects in insulin secretion. J Endocrinol 2011; 210:335-47. [PMID: 21700659 DOI: 10.1530/joe-11-0126] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Increased glucose production is associated with fasting hyperglycaemia in type 2 diabetes but whether or not it causes glucose intolerance is unclear. This study sought to determine whether a primary defect in gluconeogenesis (GNG) resulting in elevated glucose production is sufficient to induce glucose intolerance in the absence of insulin resistance and impaired insulin secretion. Progression of glucose intolerance was assessed in phosphoenolpyruvate carboxykinase (PEPCK) transgenic rats, a genetic model with a primary increase in GNG. Young (4-5 weeks of age) and adult (12-14 weeks of age) PEPCK transgenic and Piebald Virol Glaxo (PVG/c) control rats were studied. GNG, insulin sensitivity, insulin secretion and glucose tolerance were assessed by intraperitoneal and intravascular substrate tolerance tests and hyperinsulinaemic/euglycaemic clamps. Despite elevated GNG and increased glucose appearance, PEPCK transgenic rats displayed normal glucose tolerance due to adequate glucose disposal and robust glucose-mediated insulin secretion. Glucose intolerance only became apparent in the PEPCK transgenic rats following the development of insulin resistance (both hepatic and peripheral) and defective glucose-mediated insulin secretion. Taken together, a single genetic defect in GNG leading to increased glucose production does not adversely affect glucose tolerance. Insulin resistance and impaired glucose-mediated insulin secretion are required to precipitate glucose intolerance in a setting of chronic glucose oversupply.
Collapse
Affiliation(s)
- Salvatore P Mangiafico
- Department of Medicine, Heidelberg Repatriation Hospital, University of Melbourne, 300 Waterdale Road, Heidelberg Heights, Melbourne, Victoria 3081, Australia
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Lee KTY, Karunakaran S, Ho MM, Clee SM. PWD/PhJ and WSB/EiJ mice are resistant to diet-induced obesity but have abnormal insulin secretion. Endocrinology 2011; 152:3005-17. [PMID: 21673102 DOI: 10.1210/en.2011-0060] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recently, novel inbred mouse strains that are genetically distinct from the commonly used models have been developed from wild-caught mice. These wild-derived inbred strains have been included in many of the large-scale genomic projects, but their potential as models of altered obesity and diabetes susceptibility has not been assessed. We examined obesity and diabetes-related traits in response to high-fat feeding in two of these strains, PWD/PhJ (PWD) and WSB/EiJ (WSB), in comparison with C57BL/6J (B6). Young PWD mice displayed high fasting insulin levels, although they had normal insulin sensitivity. PWD mice subsequently developed a much milder and delayed-onset obesity compared with B6 mice but became as insulin resistant. PWD mice had a robust first-phase and increased second-phase glucose-stimulated insulin secretion in vivo, rendering them more glucose tolerant. WSB mice were remarkably resistant to diet-induced obesity and maintained very low fasting insulin throughout the study. WSB mice exhibited more rapid glucose clearance in response to an insulin challenge compared with B6 mice, consistent with their low percent body fat. Interestingly, in the absence of a measurable in vivo insulin secretion, glucose tolerance of WSB mice was better than B6 mice, likely due to their enhanced insulin sensitivity. Thus PWD and WSB are two obesity-resistant strains with unique insulin secretion phenotypes. PWD mice are an interesting model that dissociates hyperinsulinemia from obesity and insulin resistance, whereas WSB mice are a model of extraordinary resistance to a high-fat diet.
Collapse
Affiliation(s)
- Katie T Y Lee
- Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia, Canada V6T 1Z3
| | | | | | | |
Collapse
|
45
|
Meadows NA, Saxty B, Albury MS, Kettleborough CA, Ashcroft FM, Moore AL, Cox RD. A high-throughput assay for modulators of NNT activity in permeabilized yeast cells. ACTA ACUST UNITED AC 2011; 16:734-43. [PMID: 21602486 DOI: 10.1177/1087057111408088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Nicotinamide nucleotide transhydrogenase (NNT) mutant mice show glucose intolerance with impaired insulin secretion during glucose tolerance tests. Uncoupling of the β cell mitochondrial metabolism due to such mutations makes NNT a novel target for therapeutics in the treatment of pathologies such as type 2 diabetes. The authors propose that increasing NNT activity would help reduce deleterious buildup of reactive oxygen species in the inner mitochondrial matrix. They have expressed human Nnt cDNA for the first time in Saccharomyces cerevisiae, and transhydrogenase activity in mitochondria isolated from these cells is six times greater than is seen in wild-type mitochondria. The same mitochondria have partially uncoupled respiration, and the cells have slower growth rates compared to cells that do not express NNT. The authors have used NNT's role as a redox-driven proton pump to develop a robust fluorimetric assay in permeabilized yeast. Screening in parallel a library of known pharmacologically active compounds (National Institute of Neurological Disorders and Stroke collection) against NNT ± cells, they demonstrate a robust and reproducible assay suitable for expansion into larger and more diverse compound sets. The identification of NNT activators may help in the elucidation of the role of NNT in mammalian cells and assessing its potential as a therapeutic target for insulin secretion disorders.
Collapse
Affiliation(s)
- Nicholas A Meadows
- Medical Research Council, Mammalian Genetics Unit, Harwell Science and Innovation Campus, Oxfordshire, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
46
|
Nicholson A, Reifsnyder PC, Malcolm R, Lucas CA, MacGregor GR, Zhang W, Leiter EH. Diet-induced obesity in two C57BL/6 substrains with intact or mutant nicotinamide nucleotide transhydrogenase (Nnt) gene. Obesity (Silver Spring) 2010; 18:1902-5. [PMID: 20057372 PMCID: PMC2888716 DOI: 10.1038/oby.2009.477] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The C57BL/6J (B6/J) male mouse represents a standard for diet-induced obesity (DIO) and is unique in expressing a loss-of-function nicotinamide nucleotide transhydrogenase (Nnt) gene. This mutation was associated with a marked reduction in glucose-stimulated insulin secretion from B6/J islets in vitro and moderately impaired glucose clearance in vivo. To assess the contribution of this Nnt mutation, we compared DIO responsiveness of Nnt-mutant B6/J males to Nnt wild-type C57BL/6NJ (B6/NJ) males over a 14-week period of feeding a high-fat (60% of calories) diet. Initial mean body weights at 6 weeks did not distinguish the substrains and both substrains were DIO-sensitive. However, B6/J males outgained the B6/NJ males, with a significant 3 g higher mean body weight at 20 weeks accompanied by significant increases in both lean and fat mass. Mean nonfasting serum glucose over time was also significantly higher in B6/J males, as was impairment of glucose tolerance assessed at 8 and 20 weeks of age. Serum leptin, but not insulin, was significantly higher in B6/J males over time. Potential contributions of the wild-type Nnt gene were demonstrable on a lower fat diet (10% of calories) where a significantly greater weight gain over time by B6/NJ males was correlated with a significantly higher serum insulin. In conclusion, DIO developed in response to 60% fat feeding regardless of Nnt allele status. Contribution of the B6/J-unique Nnt mutation was most evident in response to 10% fat feeding that resulted in reduced serum insulin and weight gain compared to B6/NJ males.
Collapse
Affiliation(s)
| | | | | | - Charlotte A. Lucas
- Center for Mitochondrial and Molecular Medicine and Genetics, Dept of Developmental and Cell Biology, Developmental Biology Center, University of California, Irvine, Irvine, CA 92697-3940
| | - Grant R. MacGregor
- Center for Mitochondrial and Molecular Medicine and Genetics, Dept of Developmental and Cell Biology, Developmental Biology Center, University of California, Irvine, Irvine, CA 92697-3940
| | | | - Edward H. Leiter
- The Jackson Laboratory, Bar Harbor, ME 04609
- Author to whom correspondence should be addressed
| |
Collapse
|
47
|
Huxley L, Quirk PG, Cotton NPJ, White SA, Jackson JB. The specificity of proton-translocating transhydrogenase for nicotinamide nucleotides. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1807:85-94. [PMID: 20732298 DOI: 10.1016/j.bbabio.2010.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 08/10/2010] [Indexed: 11/29/2022]
Abstract
In its forward direction, transhydrogenase couples the reduction of NADP(+) by NADH to the outward translocation of protons across the membrane of bacteria and animal mitochondria. The enzyme has three components: dI and dIII protrude from the membrane and dII spans the membrane. Hydride transfer takes place between nucleotides bound to dI and dIII. Studies on the kinetics of a lag phase at the onset of a "cyclic reaction" catalysed by complexes of the dI and dIII components of transhydrogenase from Rhodospirillum rubrum, and on the kinetics of fluorescence changes associated with nucleotide binding, reveal two features. Firstly, the binding of NADP(+) and NADPH to dIII is extremely slow, and is probably limited by the conversion of the occluded to the open state of the complex. Secondly, dIII can also bind NAD(+) and NADH. Extrapolating to the intact enzyme this binding to the "wrong" site could lead to slip: proton translocation without change in the nucleotide redox state, which would have important consequences for bacterial and mitochondrial metabolism.
Collapse
Affiliation(s)
- Lucinda Huxley
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | | | | | | | | |
Collapse
|
48
|
Jitrapakdee S, Wutthisathapornchai A, Wallace JC, MacDonald MJ. Regulation of insulin secretion: role of mitochondrial signalling. Diabetologia 2010; 53:1019-32. [PMID: 20225132 PMCID: PMC2885902 DOI: 10.1007/s00125-010-1685-0] [Citation(s) in RCA: 221] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Accepted: 01/06/2010] [Indexed: 12/23/2022]
Abstract
Pancreatic beta cells are specialised endocrine cells that continuously sense the levels of blood sugar and other fuels and, in response, secrete insulin to maintain normal fuel homeostasis. During postprandial periods an elevated level of plasma glucose rapidly stimulates insulin secretion to decrease hepatic glucose output and promote glucose uptake into other tissues, principally muscle and adipose tissues. Beta cell mitochondria play a key role in this process, not only by providing energy in the form of ATP to support insulin secretion, but also by synthesising metabolites (anaplerosis) that can act, both intra- and extramitochondrially, as factors that couple glucose sensing to insulin granule exocytosis. ATP on its own, and possibly modulated by these coupling factors, triggers closure of the ATP-sensitive potassium channel, resulting in membrane depolarisation that increases intracellular calcium to cause insulin secretion. The metabolic imbalance caused by chronic hyperglycaemia and hyperlipidaemia severely affects mitochondrial metabolism, leading to the development of impaired glucose-induced insulin secretion in type 2 diabetes. It appears that the anaplerotic enzyme pyruvate carboxylase participates directly or indirectly in several metabolic pathways which are important for glucose-induced insulin secretion, including: the pyruvate/malate cycle, the pyruvate/citrate cycle, the pyruvate/isocitrate cycle and glutamate-dehydrogenase-catalysed alpha-ketoglutarate production. These four pathways enable 'shuttling' or 'recycling' of these intermediate(s) into and out of mitochondrion, allowing continuous production of intracellular messenger(s). The purpose of this review is to present an account of recent progress in this area of central importance in the realm of diabetes and obesity research.
Collapse
Affiliation(s)
- S Jitrapakdee
- Molecular Metabolism Research Group, Department of Biochemistry, Faculty of Science, Mahidol University, Rama 6 Road, Phya-Thai, Bangkok 10400, Thailand.
| | | | | | | |
Collapse
|
49
|
Houtkooper RH, Cantó C, Wanders RJ, Auwerx J. The secret life of NAD+: an old metabolite controlling new metabolic signaling pathways. Endocr Rev 2010; 31:194-223. [PMID: 20007326 PMCID: PMC2852209 DOI: 10.1210/er.2009-0026] [Citation(s) in RCA: 692] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A century after the identification of a coenzymatic activity for NAD(+), NAD(+) metabolism has come into the spotlight again due to the potential therapeutic relevance of a set of enzymes whose activity is tightly regulated by the balance between the oxidized and reduced forms of this metabolite. In fact, the actions of NAD(+) have been extended from being an oxidoreductase cofactor for single enzymatic activities to acting as substrate for a wide range of proteins. These include NAD(+)-dependent protein deacetylases, poly(ADP-ribose) polymerases, and transcription factors that affect a large array of cellular functions. Through these effects, NAD(+) provides a direct link between the cellular redox status and the control of signaling and transcriptional events. Of particular interest within the metabolic/endocrine arena are the recent results, which indicate that the regulation of these NAD(+)-dependent pathways may have a major contribution to oxidative metabolism and life span extension. In this review, we will provide an integrated view on: 1) the pathways that control NAD(+) production and cycling, as well as its cellular compartmentalization; 2) the signaling and transcriptional pathways controlled by NAD(+); and 3) novel data that show how modulation of NAD(+)-producing and -consuming pathways have a major physiological impact and hold promise for the prevention and treatment of metabolic disease.
Collapse
Affiliation(s)
- Riekelt H Houtkooper
- Ecole Polytechnique Fédérale de Lausanne, Laboratory for Integrative and Systems Physiology, Building AI, Station 15, CH-1015 Lausanne, Switzerland
| | | | | | | |
Collapse
|
50
|
Andrikopoulos S. Obesity and type 2 diabetes: slow down!--Can metabolic deceleration protect the islet beta cell from excess nutrient-induced damage? Mol Cell Endocrinol 2010; 316:140-6. [PMID: 19815054 DOI: 10.1016/j.mce.2009.09.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2009] [Revised: 09/08/2009] [Accepted: 09/28/2009] [Indexed: 01/09/2023]
Abstract
Islet beta-cell dysfunction is a characteristic and the main cause of hyperglycaemia of Type 2 diabetes. Understanding the mechanisms that cause beta-cell dysfunction will lead to better therapeutic outcomes for patients with Type 2 diabetes. Chronic fatty acid exposure of susceptible islet beta-cells causes dysfunction and death and this is associated with increased reactive oxygen species production leading to oxidative stress and increased endoplasmic reticulum stress. We present the hypothesis that metabolic deceleration can reduce both oxidative and endoplasmic reticulum stress and lead to improved beta-cell function and viability when exposed to a deleterious fat milieu. This is illustrated by the C57BL/6J mouse which is characterised by reduced insulin secretion and glucose intolerance associated with a mutation in nicotinamide nucleotide transhydrogenase (Nnt) but is resistant to obesity induced diabetes. On the other hand the DBA/2 mouse has comparatively higher insulin secretion and better glucose tolerance associated with increased Nnt activity but is susceptible to obesity-induced diabetes, possibly as a result of increased oxidative stress. We therefore suggest that in states of excess nutrient load, a reduced ability to metabolise this load may protect both the function and viability of beta-cells. Strategies that reduce metabolic flux when beta-cells are exposed to nutrient excess need to be considered when treating Type 2 diabetes.
Collapse
Affiliation(s)
- S Andrikopoulos
- Department of Medicine (AH/NH), University of Melbourne, Heidelberg Repatriation Hospital, Heidelberg Heights, Victoria, Australia.
| |
Collapse
|