1
|
Velagapudi S, Karsai G, Karsai M, Mohammed SA, Montecucco F, Liberale L, Lee H, Carbone F, Adami GF, Yang K, Crucet M, Stein S, Paneni F, Lapikova-Bryhinska T, Jang HD, Kraler S, Vdovenko D, Züllig RA, Camici GG, Kim HS, Laaksonen R, Gerber PA, Hornemann T, Akhmedov A, Lüscher TF. Inhibition of de novo ceramide synthesis by sirtuin-1 improves beta-cell function and glucose metabolism in type 2 diabetes. Cardiovasc Res 2024; 120:1265-1278. [PMID: 38739545 DOI: 10.1093/cvr/cvae100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/03/2024] [Accepted: 03/26/2024] [Indexed: 05/16/2024] Open
Abstract
AIMS Obesity and type 2 diabetes (T2D) are major risk factors for cardiovascular (CV) diseases. Dysregulated pro-apoptotic ceramide synthesis reduces β-cell insulin secretion, thereby promoting hyperglycaemic states that may manifest as T2D. Pro-apoptotic ceramides modulate insulin sensitivity and glucose tolerance while being linked to poor CV outcomes. Sirtuin-1 (SIRT1) is a NAD + -dependent deacetylase that protects against pancreatic β-cell dysfunction; however, systemic levels are decreased in obese-T2D mice and may promote pro-apoptotic ceramide synthesis and hyperglycaemia. Herein, we aimed to assess the effects of restoring circulating SIRT1 levels to prevent metabolic imbalance in obese and diabetic mice. METHODS AND RESULTS Circulating SIRT1 levels were reduced in obese-diabetic mice (db/db) as compared to age-matched non-diabetic db/+ controls. Restoration of SIRT1 plasma levels with recombinant murine SIRT1 for 4 weeks prevented body weight gain and improved glucose tolerance, insulin sensitivity, and vascular function in mice models of obesity and T2D. Untargeted lipidomics revealed that SIRT1 restored insulin secretory function of β-cells by reducing synthesis and accumulation of pro-apoptotic ceramides. Molecular mechanisms involved direct binding to and deacetylation of Toll-like receptor 4 (TLR4) by SIRT1 in β-cells, thereby decreasing the rate-limiting enzymes of sphingolipid synthesis SPTLC1/2 via AKT/NF-κB. Among patients with T2D, those with high baseline plasma levels of SIRT1 prior to metabolic surgery displayed restored β-cell function (HOMA2-β) and were more likely to have T2D remission during follow-up. CONCLUSION Acetylation of TLR4 promotes β-cell dysfunction via ceramide synthesis in T2D, which is blunted by systemic SIRT1 replenishment. Hence, restoration of systemic SIRT1 may provide a novel therapeutic strategy to counteract toxic ceramide synthesis and mitigate CV complications of T2D.
Collapse
Affiliation(s)
- Srividya Velagapudi
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Gergely Karsai
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland
| | - Maria Karsai
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zürich and University of Zürich, Zürich, Switzerland
| | - Shafeeq A Mohammed
- Department of Cardiology, Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital and University of Zürich, Zürich, Switzerland
| | - Fabrizio Montecucco
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa School of Medicine, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| | - Luca Liberale
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa School of Medicine, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| | - Hwan Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Federico Carbone
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa School of Medicine, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| | - Giovanni Francesco Adami
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa School of Medicine, Genoa, Italy
| | - Kangmin Yang
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Margot Crucet
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Sokrates Stein
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Franceso Paneni
- Department of Cardiology, Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital and University of Zürich, Zürich, Switzerland
| | | | - Hyun-Duk Jang
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Daria Vdovenko
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Richard Arnold Züllig
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zürich and University of Zürich, Zürich, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Hyo-Soo Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Reijo Laaksonen
- Zora Biosciences and Finnish Cardiovascular Research Center, Finland Medical School, Tampere University, Tampere, Finland
| | - Philipp A Gerber
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zürich and University of Zürich, Zürich, Switzerland
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
- Royal Brompton and Harefield Hospitals, Imperial College and King's College, London, United Kingdom
| |
Collapse
|
2
|
Siwan D, Nandave M, Gilhotra R, Almalki WH, Gupta G, Gautam RK. Unlocking β-cell restoration: The crucial role of PDX1 in diabetes therapy. Pathol Res Pract 2024; 254:155131. [PMID: 38309018 DOI: 10.1016/j.prp.2024.155131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/05/2024]
Abstract
Diabetes has been a significant healthcare problem worldwide for a considerable period. The primary objective of diabetic treatment plans is to control the symptoms associated with the pathology. To effectively combat diabetes, it is crucial to comprehend the disease's etiology, essential factors, and the relevant processes involving β-cells. The development of the pancreas, maturation, and maintenance of β-cells, and their role in regular insulin function are all regulated by PDX1. Therefore, understanding the regulation of PDX1 and its interactions with signaling pathways involved in β-cell differentiation and proliferation are crucial elements of alternative diabetes treatment strategies. The present review aims to explore the protective role of PDX1 in β-cell proliferation through signaling pathways. The main keywords chosen for this review include "PDX1 for β-cell mass," "β-cell proliferation," "β-cell restoration via PDX1," and "mechanism of PDX1 in β-cells." A comprehensive literature search was conducted using various internet search engines, such as PubMed, Science Direct, and other publication databases. We summarize several approaches to generating β-cells from alternative cell sources, employing PDX1 under various modified growth conditions and different transcriptional factors. Our analysis highlights the unique potential of PDX1 as a promising target in molecular and cell-based therapies for diabetes.
Collapse
Affiliation(s)
- Deepali Siwan
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India
| | - Mukesh Nandave
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India.
| | - Ritu Gilhotra
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Gaurav Gupta
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman, 346, United Arab Emirates
| | - Rupesh K Gautam
- Department of Pharmacology, Indore Institute of Pharmacy, IIST Campus, Opposite IIM Indore, Rau-Pithampur Road, Indore 453331, Madhya Pradesh, India
| |
Collapse
|
3
|
Lacombe J, Guo K, Bonneau J, Faubert D, Gioanni F, Vivoli A, Muir SM, Hezzaz S, Poitout V, Ferron M. Vitamin K-dependent carboxylation regulates Ca 2+ flux and adaptation to metabolic stress in β cells. Cell Rep 2023; 42:112500. [PMID: 37171959 DOI: 10.1016/j.celrep.2023.112500] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 02/24/2023] [Accepted: 04/26/2023] [Indexed: 05/14/2023] Open
Abstract
Vitamin K is a micronutrient necessary for γ-carboxylation of glutamic acids. This post-translational modification occurs in the endoplasmic reticulum (ER) and affects secreted proteins. Recent clinical studies implicate vitamin K in the pathophysiology of diabetes, but the underlying molecular mechanism remains unknown. Here, we show that mouse β cells lacking γ-carboxylation fail to adapt their insulin secretion in the context of age-related insulin resistance or diet-induced β cell stress. In human islets, γ-carboxylase expression positively correlates with improved insulin secretion in response to glucose. We identify endoplasmic reticulum Gla protein (ERGP) as a γ-carboxylated ER-resident Ca2+-binding protein expressed in β cells. Mechanistically, γ-carboxylation of ERGP protects cells against Ca2+ overfilling by diminishing STIM1 and Orai1 interaction and restraining store-operated Ca2+ entry. These results reveal a critical role of vitamin K-dependent carboxylation in regulation of Ca2+ flux in β cells and in their capacity to adapt to metabolic stress.
Collapse
Affiliation(s)
- Julie Lacombe
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada.
| | - Kevin Guo
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada; Division of Experimental Medicine, McGill University, Montréal, QC H4A 3J1, Canada
| | - Jessica Bonneau
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada; Programme de Biologie Moléculaire, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Denis Faubert
- Mass Spectrometry and Proteomics Platform, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Florian Gioanni
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Alexis Vivoli
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Sarah M Muir
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Soraya Hezzaz
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; Département de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Mathieu Ferron
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada; Division of Experimental Medicine, McGill University, Montréal, QC H4A 3J1, Canada; Programme de Biologie Moléculaire, Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada.
| |
Collapse
|
4
|
Goutchtat R, Quenon A, Clarisse M, Delalleau N, Coddeville A, Gobert M, Gmyr V, Kerr-Conte J, Pattou F, Hubert T. Effects of subtotal pancreatectomy and long-term glucose and lipid overload on insulin secretion and glucose homeostasis in minipigs. Endocrinol Diabetes Metab 2023:e425. [PMID: 37144278 DOI: 10.1002/edm2.425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 05/06/2023] Open
Abstract
INTRODUCTION Nowadays, there are no strong diabetic pig models, yet they are required for various types of diabetes research. Using cutting-edge techniques, we attempted to develop a type 2 diabetic minipig model in this study by combining a partial pancreatectomy (Px) with an energetic overload administered either orally or parenterally. METHODS Different groups of minipigs, including Göttingen-like (GL, n = 17) and Ossabaw (O, n = 4), were developed. Prior to and following each intervention, metabolic assessments were conducted. First, the metabolic responses of the Göttingen-like (n = 3) and Ossabaw (n = 4) strains to a 2-month High-Fat, High-Sucrose diet (HFHSD) were compared. Then, other groups of GL minipigs were established: with a single Px (n = 10), a Px combined with a 2-month HFHSD (n = 6), and long-term intraportal glucose and lipid infusions that were either preceded by a Px (n = 4) or not (n = 4). RESULTS After the 2-month HFHSD, there was no discernible change between the GL and O minipigs. The pancreatectomized group in GL minipigs showed a significantly lower Acute Insulin Response (AIR) (18.3 ± 10.0 IU/mL after Px vs. 34.9 ± 13.7 IU/mL before, p < .0005). In both long-term intraportal infusion groups, an increase in the Insulinogenic (IGI) and Hepatic Insulin Resistance Indexes (HIRI) was found with a decrease in the AIR, especially in the pancreatectomized group (IGI: 4.2 ± 1.9 after vs. 1.5 ± 0.8 before, p < .05; HIRI (×10-5 ): 12.6 ± 7.9 after vs. 3.8 ± 4.3 before, p < .05; AIR: 24.4 ± 13.7 µIU/mL after vs. 43.9 ± 14.5 µIU/mL before, p < .005). Regardless of the group, there was no fasting hyperglycemia. CONCLUSIONS In this study, we used pancreatectomy followed by long-term intraportal glucose and lipid infusions to develop an original minipig model with metabolic syndrome and early signs of glucose intolerance. We reaffirm the pig's usefulness as a preclinical model for the metabolic syndrome but without the fasting hyperglycemia that characterizes diabetes mellitus.
Collapse
Affiliation(s)
- Rébecca Goutchtat
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, UFR3S, U1190 - Egid, Lille, France
| | - Audrey Quenon
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, UFR3S, U1190 - Egid, Lille, France
- Univ. Lille, CHU Lille, UFR3S, Département Hospitalo-Universitaire de Recherche et d'Enseignement (Dhure), Lille, France
| | | | - Nathalie Delalleau
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, UFR3S, U1190 - Egid, Lille, France
| | - Anaïs Coddeville
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, UFR3S, U1190 - Egid, Lille, France
| | - Mathilde Gobert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, UFR3S, U1190 - Egid, Lille, France
| | - Valéry Gmyr
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, UFR3S, U1190 - Egid, Lille, France
| | - Julie Kerr-Conte
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, UFR3S, U1190 - Egid, Lille, France
| | - François Pattou
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, UFR3S, U1190 - Egid, Lille, France
| | - Thomas Hubert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, UFR3S, U1190 - Egid, Lille, France
- Univ. Lille, CHU Lille, UFR3S, Département Hospitalo-Universitaire de Recherche et d'Enseignement (Dhure), Lille, France
| |
Collapse
|
5
|
Zhang M, Yang C, Zhu M, Qian L, Luo Y, Cheng H, Geng R, Xu X, Qian C, Liu Y. Saturated fatty acids entrap PDX1 in stress granules and impede islet beta cell function. Diabetologia 2021; 64:1144-1157. [PMID: 33569632 DOI: 10.1007/s00125-021-05389-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/16/2020] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS Failure of pancreatic and duodenal homeobox factor 1 (PDX1) to localise in the nucleus of islet beta cells under high-fat diet (HFD) conditions may be an early functional defect that contributes to beta cell failure in type 2 diabetes; however, the mechanism of PDX1 intracellular mislocalisation is unclear. Stress granules (SGs) are membrane-less cytoplasmic structures formed under stress that impair nucleocytoplasmic transport by sequestering nucleocytoplasmic transport factors and components of the nuclear pore complex. In this study, we investigated the stimulators that trigger SG formation in islet beta cells and the effects of SGs on PDX1 localisation and beta cell function. METHODS The effect of palmitic acid (PA) on nucleocytoplasmic transport was investigated by using two reporters, S-tdTomato and S-GFP. SG assembly in rat insulinoma cell line INS1 cells, human islets under PA stress, and the pancreas of diet-induced obese mice was analysed using immunofluorescence and immunoblotting. SG protein components were identified through mass spectrometry. SG formation was blocked by specific inhibitors or genetic deletion of essential SG proteins, and then PDX1 localisation and beta cell function were investigated in vitro and in vivo. RESULTS We showed that saturated fatty acids (SFAs) are endogenous stressors that disrupted nucleocytoplasmic transport and stimulated SG formation in pancreatic beta cells. Using mass spectrometry approaches, we revealed that several nucleocytoplasmic transport factors and PDX1 were localised to SGs after SFA treatment, which inhibited glucose-induced insulin secretion. Furthermore, we found that SFAs induced SG formation in a phosphoinositide 3-kinase (PI3K)/eukaryotic translation initiation factor 2α (EIF2α) dependent manner. Disruption of SG assembly by PI3K/EIF2α inhibitors or genetic deletion of T cell restricted intracellular antigen 1 (TIA1) in pancreatic beta cells effectively suppressed PA-induced PDX1 mislocalisation and ameliorated HFD-mediated beta cell dysfunction. CONCLUSIONS/INTERPRETATION Our findings suggest a link between SG formation and beta cell dysfunction in the presence of SFAs. Preventing SG formation may be a potential therapeutic strategy for treating obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Mu Zhang
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Chunjie Yang
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Meng Zhu
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Li Qian
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Yan Luo
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Huimin Cheng
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Rong Geng
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaojun Xu
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.
| | - Cheng Qian
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.
| | - Yu Liu
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
6
|
Maachi H, Fergusson G, Ethier M, Brill GN, Katz LS, Honig LB, Metukuri MR, Scott DK, Ghislain J, Poitout V. HB-EGF Signaling Is Required for Glucose-Induced Pancreatic β-Cell Proliferation in Rats. Diabetes 2020; 69:369-380. [PMID: 31882563 PMCID: PMC7034189 DOI: 10.2337/db19-0643] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 12/19/2019] [Indexed: 12/19/2022]
Abstract
The molecular mechanisms of β-cell compensation to metabolic stress are poorly understood. We previously observed that nutrient-induced β-cell proliferation in rats is dependent on epidermal growth factor receptor (EGFR) signaling. The aim of this study was to determine the role of the EGFR ligand heparin-binding EGF-like growth factor (HB-EGF) in the β-cell proliferative response to glucose, a β-cell mitogen and key regulator of β-cell mass in response to increased insulin demand. We show that exposure of isolated rat and human islets to HB-EGF stimulates β-cell proliferation. In rat islets, inhibition of EGFR or HB-EGF blocks the proliferative response not only to HB-EGF but also to glucose. Furthermore, knockdown of HB-EGF in rat islets blocks β-cell proliferation in response to glucose ex vivo and in vivo in transplanted glucose-infused rats. Mechanistically, we demonstrate that HB-EGF mRNA levels are increased in β-cells in response to glucose in a carbohydrate-response element-binding protein (ChREBP)-dependent manner. In addition, chromatin immunoprecipitation studies identified ChREBP binding sites in proximity to the HB-EGF gene. Finally, inhibition of Src family kinases, known to be involved in HB-EGF processing, abrogated glucose-induced β-cell proliferation. Our findings identify a novel glucose/HB-EGF/EGFR axis implicated in β-cell compensation to increased metabolic demand.
Collapse
Affiliation(s)
- Hasna Maachi
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, University of Montreal, Montreal, Quebec, Canada
| | - Grace Fergusson
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Melanie Ethier
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Gabriel N Brill
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Liora S Katz
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Lee B Honig
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Donald K Scott
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Julien Ghislain
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Lytrivi M, Castell AL, Poitout V, Cnop M. Recent Insights Into Mechanisms of β-Cell Lipo- and Glucolipotoxicity in Type 2 Diabetes. J Mol Biol 2019; 432:1514-1534. [PMID: 31628942 DOI: 10.1016/j.jmb.2019.09.016] [Citation(s) in RCA: 267] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 09/15/2019] [Accepted: 09/16/2019] [Indexed: 12/24/2022]
Abstract
The deleterious effects of chronically elevated free fatty acid (FFA) levels on glucose homeostasis are referred to as lipotoxicity, and the concurrent exposure to high glucose may cause synergistic glucolipotoxicity. Lipo- and glucolipotoxicity have been studied for over 25 years. Here, we review the current evidence supporting the role of pancreatic β-cell lipo- and glucolipotoxicity in type 2 diabetes (T2D), including lipid-based interventions in humans, prospective epidemiological studies, and human genetic findings. In addition to total FFA quantity, the quality of FFAs (saturation and chain length) is a key determinant of lipotoxicity. We discuss in vitro and in vivo experimental models to investigate lipo- and glucolipotoxicity in β-cells and describe experimental pitfalls. Lipo- and glucolipotoxicity adversely affect many steps of the insulin production and secretion process. The molecular mechanisms underpinning lipo- and glucolipotoxic β-cell dysfunction and death comprise endoplasmic reticulum stress, oxidative stress and mitochondrial dysfunction, impaired autophagy, and inflammation. Crosstalk between these stress pathways exists at multiple levels and may aggravate β-cell lipo- and glucolipotoxicity. Lipo- and glucolipotoxicity are therapeutic targets as several drugs impact the underlying stress responses in β-cells, potentially contributing to their glucose-lowering effects in T2D.
Collapse
Affiliation(s)
- Maria Lytrivi
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium; Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Anne-Laure Castell
- CRCHUM, Montréal, QC, Canada; Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Vincent Poitout
- CRCHUM, Montréal, QC, Canada; Department of Medicine, Université de Montréal, Montréal, QC, Canada.
| | - Miriam Cnop
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium; Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
8
|
Moullé VS, Tremblay C, Castell AL, Vivot K, Ethier M, Fergusson G, Alquier T, Ghislain J, Poitout V. The autonomic nervous system regulates pancreatic β-cell proliferation in adult male rats. Am J Physiol Endocrinol Metab 2019; 317:E234-E243. [PMID: 31013146 PMCID: PMC6732465 DOI: 10.1152/ajpendo.00385.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The pancreatic β-cell responds to changes in the nutrient environment to maintain glucose homeostasis by adapting its function and mass. Nutrients can act directly on the β-cell and also indirectly through the brain via autonomic nerves innervating islets. Despite the importance of the brain-islet axis in insulin secretion, relatively little is known regarding its involvement in β-cell proliferation. We previously demonstrated that prolonged infusions of nutrients in rats provoke a dramatic increase in β-cell proliferation in part because of the direct action of nutrients. Here, we addressed the contribution of the autonomic nervous system. In isolated islets, muscarinic stimulation increased, whereas adrenergic stimulation decreased, glucose-induced β-cell proliferation. Blocking α-adrenergic receptors reversed the effect of epinephrine on glucose + nonesterified fatty acids (NEFA)-induced β-cell proliferation, whereas activation of β-adrenergic receptors was without effect. Infusion of glucose + NEFA toward the brain stimulated β-cell proliferation, and this effect was abrogated following celiac vagotomy. The increase in β-cell proliferation following peripheral infusions of glucose + NEFA was not inhibited by vagotomy or atropine treatment but was blocked by coinfusion of epinephrine. We conclude that β-cell proliferation is stimulated by parasympathetic and inhibited by sympathetic signals. Whereas glucose + NEFA in the brain stimulates β-cell proliferation through the vagus nerve, β-cell proliferation in response to systemic nutrient excess does not involve parasympathetic signals but may be associated with decreased sympathetic tone.
Collapse
Affiliation(s)
- Valentine S Moullé
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
| | - Caroline Tremblay
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
| | - Anne-Laure Castell
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
| | - Kevin Vivot
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
| | - Mélanie Ethier
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
| | - Grace Fergusson
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
| | - Thierry Alquier
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
- Department of Medicine, University of Montreal , Quebec , Canada
| | - Julien Ghislain
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center , Montreal, Quebec , Canada
- CRCHUM, Montreal, Quebec , Canada
- Department of Medicine, University of Montreal , Quebec , Canada
| |
Collapse
|
9
|
Li Z, Zhou M, Cai Z, Liu H, Zhong W, Hao Q, Cheng D, Hu X, Hou J, Xu P, Xue Y, Zhou Y, Xu T. RNA-binding protein DDX1 is responsible for fatty acid-mediated repression of insulin translation. Nucleic Acids Res 2019; 46:12052-12066. [PMID: 30295850 PMCID: PMC6294501 DOI: 10.1093/nar/gky867] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 09/14/2018] [Indexed: 01/13/2023] Open
Abstract
The molecular mechanism in pancreatic β cells underlying hyperlipidemia and insulin insufficiency remains unclear. Here, we find that the fatty acid-induced decrease in insulin levels occurs due to a decrease in insulin translation. Since regulation at the translational level is generally mediated through RNA-binding proteins, using RNA antisense purification coupled with mass spectrometry, we identify a novel insulin mRNA-binding protein, namely, DDX1, that is sensitive to palmitate treatment. Notably, the knockdown or overexpression of DDX1 affects insulin translation, and the knockdown of DDX1 eliminates the palmitate-induced repression of insulin translation. Molecular mechanism studies show that palmitate treatment causes DDX1 phosphorylation at S295 and dissociates DDX1 from insulin mRNA, thereby leading to the suppression of insulin translation. In addition, DDX1 may interact with the translation initiation factors eIF3A and eIF4B to regulate translation. In high-fat diet mice, the inhibition of insulin translation happens at an early prediabetic stage before the elevation of glucose levels. We speculate that the DDX1-mediated repression of insulin translation worsens the situation of insulin resistance and contributes to the elevation of blood glucose levels in obese animals.
Collapse
Affiliation(s)
- Zonghong Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China.,Jilin Province Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China
| | - Maoge Zhou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhaokui Cai
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongyang Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Wen Zhong
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Qiang Hao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Dongwan Cheng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Xihao Hu
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Junjie Hou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Pingyong Xu
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuanchao Xue
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yifa Zhou
- Jilin Province Key Laboratory on Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics & University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
10
|
Glavas MM, Hui Q, Tudurí E, Erener S, Kasteel NL, Johnson JD, Kieffer TJ. Early overnutrition reduces Pdx1 expression and induces β cell failure in Swiss Webster mice. Sci Rep 2019; 9:3619. [PMID: 30842440 PMCID: PMC6403421 DOI: 10.1038/s41598-019-39177-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/07/2019] [Indexed: 12/25/2022] Open
Abstract
Childhood obesity and early rapid growth increase the risk for type 2 diabetes. Such early overnutrition can be modeled in mice by reducing litter size. We investigated the effects of early overnutrition and increased dietary fat intake on β cell function in Swiss Webster mice. On a moderate-fat diet, early overnutrition accelerated weight gain and induced hyperinsulinemia in pups. Early overnutrition males exhibited higher β cell mass but reduced islet insulin content and Pdx1 expression. Males had a high diabetes incidence that was increased by early overnutrition, characterized by a progressive increase in insulin secretion as well as β cell death, indicated by histological analysis and increased circulating miR-375 levels. Females maintained normoglycemia throughout life. High-fat diet (HFD) increased diabetes incidence in males, whereas low-fat diet was completely protective. This protective effect was abolished in early overnutrition males transiently exposed to HFD in early life. Although Swiss Webster mice are not known to be diabetes-prone, the high diabetes incidence suggests an underlying genetic susceptibility that can be induced by overnutrition and increased dietary fat intake in early life. Thus, the nutritional environment in early life may impact long-term β cell function and increase diabetes risk, particularly in genetically susceptible individuals.
Collapse
Affiliation(s)
- Maria M Glavas
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Queenie Hui
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Eva Tudurí
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada.,Centro de Investigación Biomédica en Red de Diabetes y , Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Suheda Erener
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Naomi L Kasteel
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - James D Johnson
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Timothy J Kieffer
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada. .,Department of Surgery, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
11
|
GPR120 protects lipotoxicity-induced pancreatic β-cell dysfunction through regulation of PDX1 expression and inhibition of islet inflammation. Clin Sci (Lond) 2019; 133:101-116. [PMID: 30523046 DOI: 10.1042/cs20180836] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/29/2018] [Accepted: 12/05/2018] [Indexed: 12/23/2022]
Abstract
G-protein coupled receptor 120 (GPR120) has been shown to act as an omega-3 unsaturated fatty acid sensor and is involved in insulin secretion. However, the underlying mechanism in pancreatic β cells remains unclear. To explore the potential link between GPR120 and β-cell function, its agonists docosahexaenoic acid (DHA) and GSK137647A were used in palmitic acid (PA)-induced pancreatic β-cell dysfunction, coupled with GPR120 knockdown (KD) in MIN6 cells and GPR120 knockout (KO) mice to identify the underlying signaling pathways. In vitro and ex vivo treatments of MIN6 cells and islets isolated from wild-type (WT) mice with DHA and GSK137647A restored pancreatic duodenal homeobox-1 (PDX1) expression levels and β-cell function via inhibiting PA-induced elevation of proinflammatory chemokines and activation of nuclear factor κB, c-Jun amino (N)-terminal kinases1/2 and p38MAPK signaling pathways. On the contrary, these GPR120 agonism-mediated protective effects were abolished in GPR120 KD cells and islets isolated from GPR120 KO mice. Furthermore, GPR120 KO mice displayed glucose intolerance and insulin resistance relative to WT littermates, and β-cell functional related genes were decreased while inflammation was exacerbated in islets with increased macrophages in pancreas from GPR120 KO mice. DHA and GSK137647A supplementation ameliorated glucose tolerance and insulin sensitivity, as well as improved Pdx1 expression and islet inflammation in diet-induced obese WT mice, but not in GPR120 KO mice. These findings indicate that GPR120 activation is protective against lipotoxicity-induced pancreatic β-cell dysfunction, via the mediation of PDX1 expression and inhibition of islet inflammation, and that GPR120 activation may serve as a preventative and therapeutic target for obesity and diabetes.
Collapse
|
12
|
Hadjzadeh M, Alikhani V, Hosseinian S, Zarei B, Keshavarzi Z. THE EFFECT OF MELATONIN AGAINST GASTRIC OXIDATIVE STRESS AND DYSLIPIDEMIA IN STREPTOZOTOCIN-INDUCED DIABETIC RATS. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2018; 14:453-458. [PMID: 31149296 PMCID: PMC6516406 DOI: 10.4183/aeb.2018.453] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate the possible protective effects of MT against gastric oxidative stress and dyslipidemia in streptozotocin (STZ) - induced diabetic rats. METHODS Forty male Wistar rats were randomly divided into five groups: control, diabetic, MT 5 mg/kg+ STZ, MT 10 mg/kg+ STZ and MT 20 mg/kg+ STZ. STZ (60 mg/kg) was intraperitoneally (ip) injected as a single dose for diabetes induction. One week after STZ administration, MT was injected daily as ip for 14 days. The levels of malondialdehyde (MDA), total thiol and glutathione, as well as superoxide dismutase (SOD) and catalase activities were measured in gastric tissue. Serum concentrations of triglycerides (TG), total cholesterol (TC), high density lipoprotein (HDL) and low density lipoprotein (LDL) were also determined. RESULTS Serum glucose significantly increased in diabetic group compared to control group. STZ induced a significant decrease in gastric tissue levels of total thiol, glutathione, catalase and SOD activities and a significant increase in MDA concentration. In diabetic rats, serum TG, LDL and TC were significantly higher and HDL was significantly lower than in the control group. Treatment of diabetic rats with MT caused a significant increase in gastric total thiol content and glutathione concentration as well as SOD and catalase activities. Gastric MDA concentration and serum LDL, TG and TC were significantly lower in MT-treated groups when compared with diabetic group. CONCLUSION These data suggested that MT has a therapeutic effect on gastric oxidative damage and dyslipidemia induced by diabetes that possibly may be due to its antioxidant effects.
Collapse
Affiliation(s)
- M.A.R. Hadjzadeh
- Mashhad University of Medical Sciences, School of Medicine, Department of physiology, Bojnurd, Iran
- Mashhad University of Medical Sciences, Psychiatry and Behavioral Sciences Research Center, Division of Neurocognitive Sciences, Bojnurd, Iran
| | - V. Alikhani
- Mashhad University of Medical Sciences, School of Medicine, Department of physiology, Bojnurd, Iran
| | - S. Hosseinian
- Mashhad University of Medical Sciences, School of Medicine, Department of physiology, Bojnurd, Iran
- Mashhad University of Medical Sciences, Neurogenic Inflammation Research Center, Mashhad, Bojnurd, Iran
| | - B. Zarei
- Mashhad University of Medical Sciences, School of Medicine, Department of physiology, Bojnurd, Iran
| | - Z. Keshavarzi
- Mashhad University of Medical Sciences, North Khorasan University of Medical Sciences, Natural Products and Medicinal Plants Research Center, Bojnurd, Iran
| |
Collapse
|
13
|
Zabielski P, Błachnio-Zabielska AU, Wójcik B, Chabowski A, Górski J. Effect of plasma free fatty acid supply on the rate of ceramide synthesis in different muscle types in the rat. PLoS One 2017; 12:e0187136. [PMID: 29095868 PMCID: PMC5667851 DOI: 10.1371/journal.pone.0187136] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 10/13/2017] [Indexed: 01/06/2023] Open
Abstract
Ceramide is a key compound in sphingolipid metabolism. Dynamics of ceramide synthesis is important in the several biological processes, such as induction of apoptosis or insulin resistance. So far, its de novo synthesis rate was evaluated indirectly, based on the content of the compound, its intermediates and the activity of respective enzymes. The aim of the present study was to directly measure ceramide synthesis rate (FSR) in different muscle types under varied plasma FFA supply in rat with the use of [U-13C] palmitate tracer and LC/MS/MS. The experiments were carried out on male Wistar rats, divided into three groups: 1-control, 2-with elevated plasma free fatty acid (FFA) concentration by means of intralipid and heparin, 3-with reduced plasma FFA concentration by means of nicotinic acid. The stable plasma FFA concentration and plasma [U-13C] palmitate enrichment was maintained for two hours by simultaneous infusion of the tracer and the respective compounds. At the end of the experiment, samples of blood from the abdominal aorta, the heart, diaphragm, soleus and white section of the gastrocnemius were taken. Muscle sphinganine, sphingosine and ceramide content and enrichment and plasma palmitate enrichment was measured with the use of LC/MS/MS. Plasma FFA concentration and composition was measured by means of gas-liquid chromatography. Under basal conditions ceramide FSR in the heart and the diaphragm was higher than in the soleus and the white gastrocnemius. Elevation in the plasma FFA concentration increased the FSR and ceramide content in each muscle, which correlated with increased HOMA-IR. The highest FSR was noted in the heart. Reduction in the plasma FFA concentration decreased ceramide FSR in each muscle type, which was accompanied by marked reduction in HOMA-IR. It is concluded that ceramide FSR depends on both the muscle type and the plasma FFA supply and is correlated with whole body insulin sensitivity under varying plasma FFA supply.
Collapse
Affiliation(s)
- Piotr Zabielski
- Department of Physiology, Medical University of Białystok, Białystok, Poland
- Department of Medical Biology, Medical University of Białystok, Białystok, Poland
- * E-mail:
| | - Agnieszka Urszula Błachnio-Zabielska
- Department of Physiology, Medical University of Białystok, Białystok, Poland
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Białystok, Białystok, Poland
| | - Beata Wójcik
- Department of Physiology, Medical University of Białystok, Białystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Białystok, Białystok, Poland
| | - Jan Górski
- Department of Physiology, Medical University of Białystok, Białystok, Poland
- Medical Institute, Łomża State University of Applied Sciences, Łomża, Poland
| |
Collapse
|
14
|
Doliba NM, Liu Q, Li C, Chen P, Liu C, Naji A, Matschinsky FM. Inhibition of cholinergic potentiation of insulin secretion from pancreatic islets by chronic elevation of glucose and fatty acids: Protection by casein kinase 2 inhibitor. Mol Metab 2017; 6:1240-1253. [PMID: 29031723 PMCID: PMC5641685 DOI: 10.1016/j.molmet.2017.07.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/24/2017] [Accepted: 07/31/2017] [Indexed: 01/24/2023] Open
Abstract
Objectives Chronic hyperlipidemia and hyperglycemia are characteristic features of type 2 diabetes (T2DM) that are thought to cause or contribute to β-cell dysfunction by “glucolipotoxicity.” Previously we have shown that acute treatment of pancreatic islets with fatty acids (FA) decreases acetylcholine-potentiated insulin secretion. This acetylcholine response is mediated by M3 muscarinic receptors, which play a key role in regulating β-cell function. Here we examine whether chronic FA exposure also inhibits acetylcholine-potentiated insulin secretion using mouse and human islets. Methods Islets were cultured for 3 or 4 days at different glucose concentration with 0.5 mM palmitic acid (PA) or a 2:1 mixture of PA and oleic acid (OA) at 1% albumin (PA/BSA molar ratio 3.3). Afterwards, the response to glucose and acetylcholine were studied in perifusion experiments. Results FA-induced impairment of insulin secretion and Ca2+ signaling depended strongly on the glucose concentrations of the culture medium. PA and OA in combination reduced acetylcholine potentiation of insulin secretion more than PA alone, both in mouse and human islets, with no evidence of a protective role of OA. In contrast, lipotoxicity was not observed with islets cultured for 3 days in medium containing less than 1 mM glucose and a mixture of glutamine and leucine (7 mM each). High glucose and FAs reduced endoplasmic reticulum (ER) Ca2+ storage capacity; however, preserving ER Ca2+ by blocking the IP3 receptor with xestospongin C did not protect islets from glucolipotoxic effects on insulin secretion. In contrast, an inhibitor of casein kinase 2 (CK2) protected the glucose dependent acetylcholine potentiation of insulin secretion in mouse and human islets against glucolipotoxicity. Conclusions These results show that chronic FA treatment decreases acetylcholine potentiation of insulin secretion and that this effect is strictly glucose dependent and might involve CK2 phosphorylation of β-cell M3 muscarinic receptors. Glucolipotoxicity impairs acetylcholine-potentiation of insulin secretion. Glucose amplification of insulin secretion rather than triggering is damaged by FA. Inhibitor of casein kinase 2 preserved islet function against glucolipotoxicity.
Collapse
Affiliation(s)
- Nicolai M Doliba
- Department of Biochemistry and Biophysics, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA.
| | - Qin Liu
- Department of Biochemistry and Biophysics, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA
| | - Changhong Li
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA; Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Pan Chen
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Chengyang Liu
- Department of Surgery, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA
| | - Ali Naji
- Department of Surgery, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA
| | - Franz M Matschinsky
- Department of Biochemistry and Biophysics, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
15
|
Moullé VS, Vivot K, Tremblay C, Zarrouki B, Ghislain J, Poitout V. Glucose and fatty acids synergistically and reversibly promote beta cell proliferation in rats. Diabetologia 2017; 60:879-888. [PMID: 28078385 PMCID: PMC5376373 DOI: 10.1007/s00125-016-4197-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/08/2016] [Indexed: 12/31/2022]
Abstract
AIMS/HYPOTHESIS The mechanisms underlying pancreatic islet mass expansion have attracted considerable interest as potential therapeutic targets to prevent or delay the onset of type 2 diabetes. While several factors promoting beta cell proliferation have been identified, in the context of nutrient excess the roles of glucose or NEFA in relation to insulin resistance remain unclear. Here we tested the hypothesis that glucose and NEFA synergistically and reversibly promote beta cell proliferation in the context of nutrient-induced insulin resistance. METHODS Using 72 h infusions of glucose (GLU) or the oleate-enriched lipid emulsion ClinOleic (CLI), singly or in combination, we assessed beta cell proliferation, islet mass and insulin sensitivity in male Lewis rats. The effects of nutrients and endogenous circulating factors were examined in isolated and transplanted islets. Reversibility was studied 3 and 6 days after the end of the infusion. RESULTS GLU infusions modestly stimulated beta cell proliferation, CLI alone had no effect and GLU+CLI infusions markedly stimulated beta cell proliferation. Insulin sensitivity was equally decreased in GLU and GLU+CLI infusions. GLU+CLI infusions also stimulated beta cell proliferation in islets transplanted under the kidney capsule, albeit to a lesser extent compared with endogenous islets. Ex vivo, the combination of glucose and NEFA enhanced beta cell proliferation in rat and human islets independently from secreted insulin, and serum from GLU+CLI-infused rats potentiated the effect of glucose. Glucose tolerance, beta cell proliferation and islet mass were all restored to normal levels 6 days after termination of the infusion. CONCLUSIONS/INTERPRETATION Glucose and NEFA synergistically and reversibly promote beta cell proliferation in part via direct action on the beta cell and independently from secreted insulin.
Collapse
Affiliation(s)
- Valentine S Moullé
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada
- The University of Montreal Hospital Research Centre (CRCHUM), 900 Saint-Denis, Montreal, QC, H2X 0A9, Canada
| | - Kevin Vivot
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada
- The University of Montreal Hospital Research Centre (CRCHUM), 900 Saint-Denis, Montreal, QC, H2X 0A9, Canada
| | - Caroline Tremblay
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada
- The University of Montreal Hospital Research Centre (CRCHUM), 900 Saint-Denis, Montreal, QC, H2X 0A9, Canada
| | - Bader Zarrouki
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada
- The University of Montreal Hospital Research Centre (CRCHUM), 900 Saint-Denis, Montreal, QC, H2X 0A9, Canada
| | - Julien Ghislain
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada
- The University of Montreal Hospital Research Centre (CRCHUM), 900 Saint-Denis, Montreal, QC, H2X 0A9, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada.
- The University of Montreal Hospital Research Centre (CRCHUM), 900 Saint-Denis, Montreal, QC, H2X 0A9, Canada.
- Department of Medicine, University of Montreal, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC, Canada.
| |
Collapse
|
16
|
Bergeron V, Ghislain J, Poitout V. The P21-activated kinase PAK4 is implicated in fatty-acid potentiation of insulin secretion downstream of free fatty acid receptor 1. Islets 2016; 8:157-164. [PMID: 27700527 PMCID: PMC5161145 DOI: 10.1080/19382014.2016.1243191] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Free fatty acid receptor 1 (FFA1/GPR40) plays a key role in the potentiation of glucose-stimulated insulin secretion by fatty acids in pancreatic β cells. We previously demonstrated that GPR40 signaling leads to cortical actin remodeling and potentiates the second phase of insulin secretion. In this study, we examined the role of p21 activated kinase 4 (PAK4), a known regulator of cytoskeletal dynamics, in GPR40-dependent potentiation of insulin secretion. The fatty acid oleate induced PAK4 phosphorylation in human islets, in isolated mouse islets and in the insulin secreting cell line INS832/13. However, oleate-induced PAK4 phosphorylation was not observed in GPR40-null mouse islets. siRNA-mediated knockdown of PAK4 in INS832/13 cells abrogated the potentiation of insulin secretion by oleate, whereas PAK7 knockdown had no effect. Our results indicate that PAK4 plays an important role in the potentiation of insulin secretion by fatty acids downstream of GPR40.
Collapse
Affiliation(s)
- Valérie Bergeron
- Montreal Diabetes Research Center, CRCHUM, QC, Canada
- Department of Medicine, University of Montreal, QC, Canada
| | | | - Vincent Poitout
- Montreal Diabetes Research Center, CRCHUM, QC, Canada
- Department of Medicine, University of Montreal, QC, Canada
- CONTACT Vincent Poitout CRCHUM, 900 Saint-Denis Street, Montreal, QC, Canada, H2X 0A9
| |
Collapse
|
17
|
Zhang B, Lai G, Wu J, Sun R, Xu R, Yang X, Qi Y, Zhao Y. 20-HETE attenuates the response of glucose-stimulated insulin secretion through the AKT/GSK-3β/Glut2 pathway. Endocrine 2016; 54:371-382. [PMID: 27568038 DOI: 10.1007/s12020-016-1031-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 06/20/2016] [Indexed: 12/29/2022]
Abstract
We previously generated cytochrome P450 4F2 (CYP4F2) transgenic mice that have high levels of 20-hydroxyeicosatetraenoic acid (20-HETE) production; these mice exhibit both hypertension and hyperglycemia without insulin resistance. Currently, it is unclear whether and how 20-HETE affects insulin secretion, thus resulting in hyperglycemia. In this study, we found that 20-HETE attenuated glucose-stimulated insulin secretion (GSIS) in CYP4F2 transgenic mice as well as in rat insulinoma INS-1E cells treated with 0.5 μM 20-HETE. HET0016, a selective inhibitor of 20-HETE synthesis, reversed the reduction in GSIS leading to a decrease in blood glucose in the transgenic mice. Furthermore, the expression of glucose transporter 2 (Glut2), Ser473 phosphorylation of protein kinase B (AKT), and Ser9 phosphorylation of glycogen synthase kinase-3β (GSK-3β) were decreased in CYP4F2 transgenic mice compared with wild-type mice. In vitro experiments in INS-1E cells revealed that 20-HETE activated the AKT/GSK-3β pathway and thereby decreased Glut2 expression by inhibiting activator protein 1 (AP-1). TWS119, a GSK-3β selective inhibitor, blocked the 20-HETE-mediated reduction in Glut2 expression. Therefore, we concluded that 20-HETE inhibition of Glut2 contributes to the reduction in GSIS, at least in part, through the AKT/GSK-3β/AP-1/Glut2 pathway.
Collapse
Affiliation(s)
- Bijun Zhang
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Medical Genetics, China Medical University, Shenyang, Liaoning, China
| | - Guangrui Lai
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jingjing Wu
- Department of Medical Genetics, China Medical University, Shenyang, Liaoning, China
| | - Ru Sun
- Department of Medical Genetics, China Medical University, Shenyang, Liaoning, China
| | - Runhong Xu
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Medical Genetics, China Medical University, Shenyang, Liaoning, China
| | - Xianghong Yang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yafei Qi
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yanyan Zhao
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
- Department of Medical Genetics, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
18
|
Ghislain J, Fontés G, Tremblay C, Kebede MA, Poitout V. Dual-Reporter β-Cell-Specific Male Transgenic Rats for the Analysis of β-Cell Functional Mass and Enrichment by Flow Cytometry. Endocrinology 2016; 157:1299-306. [PMID: 26671180 PMCID: PMC4769371 DOI: 10.1210/en.2015-1550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mouse β-cell-specific reporter lines have played a key role in diabetes research. Although the rat provides several advantages, its use has lagged behind the mouse due to the relative paucity of genetic models. In this report we describe the generation and characterization of transgenic rats expressing a Renilla luciferase (RLuc)-enhanced yellow fluorescent protein (YFP) fusion under control of a 9-kb genomic fragment from the rat ins2 gene (RIP7-RLuc-YFP). Analysis of RLuc luminescence and YFP fluorescence revealed that reporter expression is restricted to β-cells in the adult rat. Physiological characteristics including body weight, fat and lean mass, fasting and fed glucose levels, glucose and insulin tolerance, and β-cell mass were similar between two RIP7-RLuc-YFP lines and wild-type littermates. Glucose-induced insulin secretion in isolated islets was indistinguishable from controls in one of the lines, whereas surprisingly, insulin secretion was defective in the second line. Consequently, subsequent studies were limited to the former line. We asked whether transgene activity was responsive to glucose as shown previously for the ins2 gene. Exposing islets ex vivo to high glucose (16.7 mM) or in vivo infusion of glucose for 24 hours increased luciferase activity in islets, whereas the fraction of YFP-positive β-cells after glucose infusion was unchanged. Finally, we showed that fluorescence-activated cell sorting of YFP-positive islet cells can be used to enrich for β-cells. Overall, this transgenic line will enable for the first time the application of both fluorescence and bioluminescence/luminescence-based approaches for the study of rat β-cells.
Collapse
Affiliation(s)
- Julien Ghislain
- Montreal Diabetes Research Center (J.G., G.F., C.T., M.A.K., V.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (J.G., G.F., C.T., M.A.K., V.P.), and Departments of Medicine (V.P.) and Biochemistry (V.P.), University of Montreal, Montréal, Québec, Canada H2X 0A9
| | - Ghislaine Fontés
- Montreal Diabetes Research Center (J.G., G.F., C.T., M.A.K., V.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (J.G., G.F., C.T., M.A.K., V.P.), and Departments of Medicine (V.P.) and Biochemistry (V.P.), University of Montreal, Montréal, Québec, Canada H2X 0A9
| | - Caroline Tremblay
- Montreal Diabetes Research Center (J.G., G.F., C.T., M.A.K., V.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (J.G., G.F., C.T., M.A.K., V.P.), and Departments of Medicine (V.P.) and Biochemistry (V.P.), University of Montreal, Montréal, Québec, Canada H2X 0A9
| | - Melkam A Kebede
- Montreal Diabetes Research Center (J.G., G.F., C.T., M.A.K., V.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (J.G., G.F., C.T., M.A.K., V.P.), and Departments of Medicine (V.P.) and Biochemistry (V.P.), University of Montreal, Montréal, Québec, Canada H2X 0A9
| | - Vincent Poitout
- Montreal Diabetes Research Center (J.G., G.F., C.T., M.A.K., V.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (J.G., G.F., C.T., M.A.K., V.P.), and Departments of Medicine (V.P.) and Biochemistry (V.P.), University of Montreal, Montréal, Québec, Canada H2X 0A9
| |
Collapse
|
19
|
Brajkovic S, Ferdaoussi M, Pawlowski V, Ezanno H, Plaisance V, Zmuda E, Hai T, Annicotte JS, Waeber G, Abderrahmani A. Islet Brain 1 Protects Insulin Producing Cells against Lipotoxicity. J Diabetes Res 2016; 2016:9158562. [PMID: 26665154 PMCID: PMC4655268 DOI: 10.1155/2016/9158562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 03/06/2015] [Indexed: 01/09/2023] Open
Abstract
Chronic intake of saturated free fatty acids is associated with diabetes and may contribute to the impairment of functional beta cell mass. Mitogen activated protein kinase 8 interacting protein 1 also called islet brain 1 (IB1) is a candidate gene for diabetes that is required for beta cell survival and glucose-induced insulin secretion (GSIS). In this study we investigated whether IB1 expression is required for preserving beta cell survival and function in response to palmitate. Chronic exposure of MIN6 and isolated rat islets cells to palmitate led to reduction of the IB1 mRNA and protein content. Diminution of IB1 mRNA and protein level relied on the inducible cAMP early repressor activity and proteasome-mediated degradation, respectively. Suppression of IB1 level mimicked the harmful effects of palmitate on the beta cell survival and GSIS. Conversely, ectopic expression of IB1 counteracted the deleterious effects of palmitate on the beta cell survival and insulin secretion. These findings highlight the importance in preserving the IB1 content for protecting beta cell against lipotoxicity in diabetes.
Collapse
Affiliation(s)
- Saška Brajkovic
- Service of Internal Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland
- University of Lille, European Genomic Institute for Diabetes (EGID) FR 3508, UMR CNRS 8199, Faculty of Medicine West, 1 place de Verdun, 59045 Lille, France
| | - Mourad Ferdaoussi
- Service of Internal Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland
- University of Lille, European Genomic Institute for Diabetes (EGID) FR 3508, UMR CNRS 8199, Faculty of Medicine West, 1 place de Verdun, 59045 Lille, France
- Department of Pharmacology and the Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Valérie Pawlowski
- University of Lille, European Genomic Institute for Diabetes (EGID) FR 3508, UMR CNRS 8199, Faculty of Medicine West, 1 place de Verdun, 59045 Lille, France
- University of Lille, EGID FR 3508, Department of Endocrine Surgery, Lille University Hospital, UMR INSERM 1190, Lille, France
| | - Hélène Ezanno
- University of Lille, European Genomic Institute for Diabetes (EGID) FR 3508, UMR CNRS 8199, Faculty of Medicine West, 1 place de Verdun, 59045 Lille, France
| | - Valérie Plaisance
- University of Lille, European Genomic Institute for Diabetes (EGID) FR 3508, UMR CNRS 8199, Faculty of Medicine West, 1 place de Verdun, 59045 Lille, France
| | - Erik Zmuda
- Department of Molecular and Cellular Biochemistry, Ohio State University, 1060 Carmack Road, Columbus, OH, USA
| | - Tsonwin Hai
- Department of Molecular and Cellular Biochemistry, Ohio State University, 1060 Carmack Road, Columbus, OH, USA
| | - Jean-Sébastien Annicotte
- University of Lille, European Genomic Institute for Diabetes (EGID) FR 3508, UMR CNRS 8199, Faculty of Medicine West, 1 place de Verdun, 59045 Lille, France
| | - Gérard Waeber
- Service of Internal Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland
| | - Amar Abderrahmani
- University of Lille, European Genomic Institute for Diabetes (EGID) FR 3508, UMR CNRS 8199, Faculty of Medicine West, 1 place de Verdun, 59045 Lille, France
- *Amar Abderrahmani:
| |
Collapse
|
20
|
Tabara Y, Takahashi Y, Setoh K, Kawaguchi T, Gotoh N, Terao C, Yamada R, Kosugi S, Sekine A, Nakayama T, Matsuda F. Synergistic association of elevated serum free fatty acid and glucose levels with large arterial stiffness in a general population: The Nagahama Study. Metabolism 2016; 65:66-72. [PMID: 26683797 DOI: 10.1016/j.metabol.2015.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/14/2015] [Accepted: 10/03/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Previous studies have reported that artificial increases in circulating free fatty acid (FFA) levels might have adverse effects on the vasculature. However, whether or not this effect can be extrapolated to physiological variations in FFA levels has not been clarified. Given that FFAs exert a lipotoxic effect on pancreatic β-cells and might directly damage the arterial endothelium, we hypothesized that these adverse effects might synergize with hyperglycemia. METHODS A total of 9396 Japanese subjects were included in the study. Serum FFA levels were measured at baseline examination. Brachial-to-ankle pulse wave velocity (baPWV) was measured as an index of arterial stiffness. RESULTS As serum levels of FFA were markedly lower in subjects with higher insulin level, a significant association between FFA levels and baPWV was observed only in subjects with blood samples taken under fasting (≥12 h, P<0.001) or near-fasting (5-11 h, P<0.001) conditions, and not in those taken under non-fasting (<5 h, P=0.307) conditions. Although type 2 diabetes and HbA1c showed a strong association with baPWV, the association between FFA level and baPWV remained significant (β=0.052, P<0.001) after adjustment for glycemic levels. In addition to their direct relationship, FFA and glucose levels were synergistically associated with baPWV (FFA(⁎)glucose; β=0.036, P<0.001). Differences in baPWV between the lowest and highest subgroups divided by a combination of FFA and glucose reached approximately 300 cm/s. CONCLUSIONS Physiological variations in FFA concentrations might be a risk factor for large arterial stiffness. FFA and hyperglycemia exert a synergistic adverse effect on the vasculature.
Collapse
Affiliation(s)
- Yasuharu Tabara
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Yoshimitsu Takahashi
- Department of Health Informatics, Kyoto University School of Public Health, Kyoto, Japan
| | - Kazuya Setoh
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Norimoto Gotoh
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Chikashi Terao
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryo Yamada
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shinji Kosugi
- Department of Medical Ethics and Medical Genetics, Kyoto University School of Public Health, Kyoto, Japan
| | - Akihiro Sekine
- Center for Preventive Medical Science, Chiba University, Chiba Japan
| | - Takeo Nakayama
- Department of Health Informatics, Kyoto University School of Public Health, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
21
|
Farsi PF, Djazayery A, Eshraghian MR, Koohdani F, Saboor-Yaraghi AA, Derakhshanian H, Zarei M, Javanbakht MH, Djalali M. Effects of supplementation with omega-3 on insulin sensitivity and non-esterified free fatty acid (NEFA) in type 2 diabetic patients. ACTA ACUST UNITED AC 2015; 58:335-40. [PMID: 24936727 DOI: 10.1590/0004-2730000002861] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/24/2013] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The aim of this study was to determine the role of omega-3 supplementation on NEFA concentration, insulin sensitivity and resistance, and glucose and lipid metabolism in type 2 diabetic patients. SUBJECTS AND METHODS Forty-four type 2 diabetic patients were randomly recruited into two groups. Group A received 4 g/day omega-3 soft gels, and group B received a placebo for 10 wks. Blood samples were collected after 12-h fast. Physical activity records, three-day food records, and anthropometric measurements were obtained from all participants at the beginning and end of the study. RESULTS Omega-3 supplementation caused a significant reduction in NEFA in the intervention group compared with the placebo group (P = 0.009). Additionally, the administration of omega-3 resulted in significantly greater changes (Diff) for the intervention group in various parameters, such as insulin and Quicki indices compared with the placebo group (P < 0.05). CONCLUSIONS Omega-3 fatty acid supplementation in type 2 diabetic patients improved insulin sensitivity, probably due to the decrease in NEFA concentrations.
Collapse
Affiliation(s)
- Payam Farahbakhsh Farsi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolghassem Djazayery
- Department of Nutrition and Biochemistry, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Eshraghian
- Department of Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Fariba Koohdani
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Akbar Saboor-Yaraghi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Hoda Derakhshanian
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahnaz Zarei
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hassan Javanbakht
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Djalali
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Erion KA, Berdan CA, Burritt NE, Corkey BE, Deeney JT. Chronic Exposure to Excess Nutrients Left-shifts the Concentration Dependence of Glucose-stimulated Insulin Secretion in Pancreatic β-Cells. J Biol Chem 2015; 290:16191-201. [PMID: 25934392 DOI: 10.1074/jbc.m114.620351] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Indexed: 01/21/2023] Open
Abstract
Hyperinsulinemia (HI) is elevated plasma insulin at basal glucose. Impaired glucose tolerance is associated with HI, although the exact cause and effect relationship remains poorly defined. We tested the hypothesis that HI can result from an intrinsic response of the β-cell to chronic exposure to excess nutrients, involving a shift in the concentration dependence of glucose-stimulated insulin secretion. INS-1 (832/13) cells were cultured in either a physiological (4 mm) or high (11 mm) glucose concentration with or without concomitant exposure to oleate. Isolated rat islets were also cultured with or without oleate. A clear hypersensitivity to submaximal glucose concentrations was evident in INS-1 cells cultured in excess nutrients such that the 25% of maximal (S0.25) glucose-stimulated insulin secretion was significantly reduced in cells cultured in 11 mm glucose (S0.25 = 3.5 mm) and 4 mm glucose with oleate (S0.25 = 4.5 mm) compared with 4 mm glucose alone (S0.25 = 5.7 mm). The magnitude of the left shift was linearly correlated with intracellular lipid stores in INS-1 cells (r(2) = 0.97). We observed no significant differences in the dose responses for glucose stimulation of respiration, NAD(P)H autofluorescence, or Ca(2+) responses between left- and right-shifted β-cells. However, a left shift in the sensitivity of exocytosis to Ca(2+) was documented in permeabilized INS-1 cells cultured in 11 versus 4 mm glucose (S0.25 = 1.1 and 1.7 μm, respectively). Our results suggest that the sensitivity of exocytosis to triggering is modulated by a lipid component, the levels of which are influenced by the culture nutrient environment.
Collapse
Affiliation(s)
- Karel A Erion
- From the Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Charles A Berdan
- From the Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Nathan E Burritt
- From the Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Barbara E Corkey
- From the Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Jude T Deeney
- From the Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| |
Collapse
|
23
|
Lee HJ, Choi YJ, Park SY, Kim JY, Won KC, Son JK, Kim YW. Hexane Extract of Orthosiphon stamineus Induces Insulin Expression and Prevents Glucotoxicity in INS-1 Cells. Diabetes Metab J 2015; 39:51-8. [PMID: 25729713 PMCID: PMC4342537 DOI: 10.4093/dmj.2015.39.1.51] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 06/02/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Hyperglycemia, a characteristic feature of diabetes, induces glucotoxicity in pancreatic β-cells, resulting in further impairment of insulin secretion and worsening glycemic control. Thus, preservation of insulin secretory capacity is essential for the management of type 2 diabetes. In this study, we evaluated the ability of an Orthosiphon stamineus (OS) extract to prevent glucotoxicity in insulin-producing cells. METHODS We measured insulin mRNA expression and glucose-stimulated insulin secretion (GSIS) in OS-treated INS-1 cells after exposure to a high glucose (HG; 30 mM) concentration. RESULTS The hexane extract of OS elevated mRNA expression of insulin as well as pancreatic and duodenal homeobox-1 of INS-1 cells in a dose-dependent manner. The hexane OS extract also increased the levels of phosphorylated phosphatidylinositol 3-kinase (PI3K) in a concentration-dependent manner. Additionally, Akt phosphorylation was elevated by treatment with 100 and 200 µmol of the hexane OS extract. Three days of HG exposure suppressed insulin mRNA expression and GSIS; these expressions were restored by treatment with the hexane OS extract. HG elevated peroxide levels in the INS-1 cells. These levels were unaffected by OS treatment under both normal and hyperglycemic conditions. CONCLUSION Our results suggested that the hexane extract of OS elevates insulin mRNA expression and prevents glucotoxicity induced by a 3-day treatment with HG. This was associated with the activation of PI-3K and Akt.
Collapse
Affiliation(s)
- Hae-Jung Lee
- Department of Pediatrics, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Yoon-Jung Choi
- Department of Physiology, Yeungnam University College of Medicine, Daegu, Korea
| | - So-Young Park
- Department of Physiology, Yeungnam University College of Medicine, Daegu, Korea
| | - Jong-Yeon Kim
- Department of Physiology, Yeungnam University College of Medicine, Daegu, Korea
| | - Kyu-Chang Won
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Jong-Keun Son
- Yeungnam University College of Pharmacy, Gyeongsan, Korea
| | - Yong-Woon Kim
- Department of Physiology, Yeungnam University College of Medicine, Daegu, Korea
| |
Collapse
|
24
|
Abstract
Over 200 million people worldwide suffer from diabetes, a disorder of glucose homeostasis. The majority of these individuals are diagnosed with type 2 diabetes. It has traditionally been thought that tissue resistance to the action of insulin is the primary defect in type 2 diabetes. However, recent longitudinal and genome‐wide association studies have shown that insulin resistance is more likely to be a precondition, and that the failure of the pancreatic β cell to meet the increased insulin requirements is the triggering factor in the development of type 2 diabetes. A major emphasis in diabetes research has therefore shifted to understanding the causes of β cell failure. Collectively, these studies have implicated a complex network of triggers, which activate intersecting execution pathways leading to β cell dysfunction and death. In the present review, we discuss these triggers (glucotoxicity, lipotoxicity, amyloid and cytokines) with respect to the pathways they activate (oxidative stress, inflammation and endoplasmic reticulum stress) and propose a model for understanding β cell failure in type 2 diabetes. (J Diabetes Invest, doi: 10.1111/j.2040‐1124.2010.00021.x, 2010)
Collapse
Affiliation(s)
- Takeshi Ogihara
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research
| | - Raghavendra G Mirmira
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research ; Departments of Medicine and Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
25
|
Semache M, Ghislain J, Zarrouki B, Tremblay C, Poitout V. Pancreatic and duodenal homeobox-1 nuclear localization is regulated by glucose in dispersed rat islets but not in insulin-secreting cell lines. Islets 2014; 6:e982376. [PMID: 25437380 PMCID: PMC4588559 DOI: 10.4161/19382014.2014.982376] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The transcription factor Pancreatic and Duodenal Homeobox-1 (PDX-1) plays a major role in the development and function of pancreatic β-cells and its mutation results in diabetes. In adult β-cells, glucose stimulates transcription of the insulin gene in part by regulating PDX-1 expression, stability and activity. Glucose is also thought to modulate PDX-1 nuclear translocation but in vitro studies examining nucleo-cytoplasmic shuttling of endogenous or ectopically expressed PDX-1 in insulin-secreting cell lines have led to conflicting results. Here we show that endogenous PDX-1 undergoes translocation from the cytoplasm to the nucleus in response to glucose in dispersed rat islets but not in insulin-secreting MIN6, HIT-T15, or INS832/13 cells. Interestingly, however, we found that a PDX-1-GFP fusion protein can shuttle from the cytoplasm to the nucleus in response to glucose stimulation in HIT-T15 cells. Our results suggest that the regulation of endogenous PDX-1 sub-cellular localization by glucose is observed in primary islets and that care should be taken when interpreting data from insulin-secreting cell lines.
Collapse
Key Words
- ANOVA, analysis of variance
- BSA, bovine serum albumin
- DAPI, 4′, 6-diamidino-2-phenylindole
- DMEM, dulbecco's modified eagle medium
- EDTA, ethylenediaminetetraacetic acid
- GFP, green fluorescent protein
- HDAC, histone deacetylase
- HIT-T15
- INS832/13
- KRBH, krebs ringer bicarbonate hepes
- MIN6
- MODY, maturity-onset diabetes of the young
- PDX-1
- PDX-1, pancreatic and duodenal homeobox-1
- SEM, standard error of the mean
- SUMO, small ubiquitin-like modifier
- T2D, type 2 diabetes
- ZDF, zucker diabetic fatty
- glucose
- glucose-stimulated insulin secretion
- nucleo-cytoplasmic shuttling
- pancreatic β cells
Collapse
Affiliation(s)
- Meriem Semache
- Montreal Diabetes Research Center; CRCHUM; Montreal, QC, Canada
- Department of Biochemistry; University of Montreal; QC, Canada
| | - Julien Ghislain
- Montreal Diabetes Research Center; CRCHUM; Montreal, QC, Canada
| | - Bader Zarrouki
- Montreal Diabetes Research Center; CRCHUM; Montreal, QC, Canada
- Department of Medicine; University of Montreal; QC, Canada
| | | | - Vincent Poitout
- Montreal Diabetes Research Center; CRCHUM; Montreal, QC, Canada
- Department of Biochemistry; University of Montreal; QC, Canada
- Department of Medicine; University of Montreal; QC, Canada
- Correspondence to: Vincent Poitout;
| |
Collapse
|
26
|
d'Assignies G, Fontés G, Kauffmann C, Latour M, Gaboury L, Boulanger Y, Van Beers BE, Soulez G, Poitout V, Tang A. Early detection of liver steatosis by magnetic resonance imaging in rats infused with glucose and intralipid solutions and correlation to insulin levels. Metabolism 2013; 62:1850-7. [PMID: 24035445 PMCID: PMC3914148 DOI: 10.1016/j.metabol.2013.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 07/25/2013] [Accepted: 08/09/2013] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Magnetic resonance (MR) techniques allow noninvasive fat quantification. We aimed to investigate the accuracy of MR imaging (MRI), MR spectroscopy (MRS) and histological techniques to detect early-onset liver steatosis in three rat phenotypes assigned to an experimental glucolipotoxic model or a control group. MATERIALS AND METHODS This study was approved by the institutional committee for the protection of animals. Thirty-two rats (13 young Wistar, 6 old Wistar and 13 diabetic Goto-Kakizaki rats) fed a standard diet were assigned to a 72h intravenous infusion of glucose and Intralipid fat emulsion or a saline infusion. Plasma insulin levels were measured. Steatosis was quantified in ex vivo livers with gradient-recalled multi-echo MRI, MRS and histology as fat fractions (FF). RESULTS A significant correlation was found between multi-echo MRI-FF and MRS-FF (r=0.81, p<0.01) and a weaker correlation was found between histology and MRS-FF (r=0.60, p<0.01). MRS and MRI accurately distinguished young Wistar and Goto-Kakizaki rats receiving the glucose+Intralipid infusion from those receiving the saline control whereas histology did not. Significant correlations were found between MRI or MRS and insulin plasma level (r=0.63, p<0.01; r=0.57, p<0.01), and between MRI or MRS and C-peptide concentration (r=0.54, p<0.01; r=0.44, p<0.02). CONCLUSIONS Multi-echo MRI and MRS may be more sensitive to measure early-onset liver steatosis than histology in an experimental glucolipotoxic rat model.
Collapse
Affiliation(s)
- Gaspard d'Assignies
- Department of Radiology, University of Montreal, Hôpital Saint-Luc, 1058 rue Saint-Denis, Montreal, Quebec, Canada, H2X 3J4
- Research Center CHUM, Hôpital Saint-Luc, 264, René-Lévesque Blvd. East, Montreal, Quebec, Canada, H2X 1P1
- Department of Radiology, Beaujon Hospital, Université Paris VII, 100 Bd du Général Leclerc, 92118 Clichy, France
| | - Ghislaine Fontés
- Research Center CHUM, Hôpital Saint-Luc, 264, René-Lévesque Blvd. East, Montreal, Quebec, Canada, H2X 1P1
- Montreal Diabetes Research Center, CRCHUM, Technopôle Angus, 2901, Rachel Street East – Room 303, Montreal, Quebec, Canada, H1W 4A4
- Department of Medicine, University of Montreal, Qc, Canada, PO Box 6128, Station Centre-ville, Montreal, Quebec, Canada, H3C 3J7
| | - Claude Kauffmann
- Department of Radiology, University of Montreal, Hôpital Saint-Luc, 1058 rue Saint-Denis, Montreal, Quebec, Canada, H2X 3J4
- Research Center CHUM, Hôpital Saint-Luc, 264, René-Lévesque Blvd. East, Montreal, Quebec, Canada, H2X 1P1
| | - Martin Latour
- Research Center CHUM, Hôpital Saint-Luc, 264, René-Lévesque Blvd. East, Montreal, Quebec, Canada, H2X 1P1
- Montreal Diabetes Research Center, CRCHUM, Technopôle Angus, 2901, Rachel Street East – Room 303, Montreal, Quebec, Canada, H1W 4A4
| | - Louis Gaboury
- Department of anatomo-pathology, Centre hospitalier de l'Université de Montréal (CHUM), 3840 rue St-Urbain, Montreal, Quebec, Canada, H2W 1T8
| | - Yvan Boulanger
- Department of Radiology, University of Montreal, Hôpital Saint-Luc, 1058 rue Saint-Denis, Montreal, Quebec, Canada, H2X 3J4
- Research Center CHUM, Hôpital Saint-Luc, 264, René-Lévesque Blvd. East, Montreal, Quebec, Canada, H2X 1P1
| | - Bernard E. Van Beers
- Department of Radiology, Beaujon Hospital, Université Paris VII, 100 Bd du Général Leclerc, 92118 Clichy, France
| | - Gilles Soulez
- Department of Radiology, University of Montreal, Hôpital Notre-Dame, 1560 rue Sherbrooke Est, Montreal, Quebec, Canada, H2L 4M1
| | - Vincent Poitout
- Research Center CHUM, Hôpital Saint-Luc, 264, René-Lévesque Blvd. East, Montreal, Quebec, Canada, H2X 1P1
- Montreal Diabetes Research Center, CRCHUM, Technopôle Angus, 2901, Rachel Street East – Room 303, Montreal, Quebec, Canada, H1W 4A4
- Department of Medicine, University of Montreal, Qc, Canada, PO Box 6128, Station Centre-ville, Montreal, Quebec, Canada, H3C 3J7
- Department of Nutrition, University of Montreal, Qc, Canada, PO Box 6128, Station Centre-ville, Montreal, Quebec, Canada, H3C 3J7
- Department of Biochemistry, University of Montreal, Qc, Canada, PO Box 6128, Station Centre-ville, Montreal, Quebec, Canada, H3C 3J7
| | - An Tang
- Department of Radiology, University of Montreal, Hôpital Saint-Luc, 1058 rue Saint-Denis, Montreal, Quebec, Canada, H2X 3J4
- Research Center CHUM, Hôpital Saint-Luc, 264, René-Lévesque Blvd. East, Montreal, Quebec, Canada, H2X 1P1
| |
Collapse
|
27
|
Fergusson G, Ethier M, Zarrouki B, Fontés G, Poitout V. A model of chronic nutrient infusion in the rat. J Vis Exp 2013. [PMID: 23979115 DOI: 10.3791/50267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Chronic exposure to excessive levels of nutrients is postulated to affect the function of several organs and tissues and to contribute to the development of the many complications associated with obesity and the metabolic syndrome, including type 2 diabetes. To study the mechanisms by which excessive levels of glucose and fatty acids affect the pancreatic beta-cell and the secretion of insulin, we have established a chronic nutrient infusion model in the rat. The procedure consists of catheterizing the right jugular vein and left carotid artery under general anesthesia; allowing a 7-day recuperation period; connecting the catheters to the pumps using a swivel and counterweight system that enables the animal to move freely in the cage; and infusing glucose and/or Intralipid (a soybean oil emulsion which generates a mixture of approximately 80% unsaturated/20% saturated fatty acids when infused with heparin) for 72 hr. This model offers several advantages, including the possibility to finely modulate the target levels of circulating glucose and fatty acids; the option to co-infuse pharmacological compounds; and the relatively short time frame as opposed to dietary models. It can be used to examine the mechanisms of nutrient-induced dysfunction in a variety of organs and to test the effectiveness of drugs in this context.
Collapse
|
28
|
Mancini AD, Poitout V. The fatty acid receptor FFA1/GPR40 a decade later: how much do we know? Trends Endocrinol Metab 2013; 24:398-407. [PMID: 23631851 DOI: 10.1016/j.tem.2013.03.003] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 03/18/2013] [Accepted: 03/21/2013] [Indexed: 11/18/2022]
Abstract
Glucose homeostasis requires the highly coordinated regulation of insulin secretion by pancreatic β cells. This is primarily mediated by glucose itself, but other nutrients, including free fatty acids (FFAs), potentiate the insulinotropic capacity of glucose. A decade ago, the seven-transmembrane domain receptor (7TMR) GPR40 was demonstrated to be predominantly expressed in β cells and activated by long-chain FFAs. This discovery added a new dimension to our understanding of FFA-mediated control of glucose homeostasis. Furthermore, GPR40 has drawn considerable interest as a novel therapeutic target to enhance insulin secretion in type 2 diabetes. However, our understanding of the biology of GPR40 remains incomplete and its physiological role controversial. Here we summarize the current state of knowledge and emerging concepts regarding the role of GPR40 in regulating glucose homeostasis.
Collapse
Affiliation(s)
- Arturo D Mancini
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | | |
Collapse
|
29
|
Semache M, Zarrouki B, Fontés G, Fogarty S, Kikani C, Chawki MB, Rutter J, Poitout V. Per-Arnt-Sim kinase regulates pancreatic duodenal homeobox-1 protein stability via phosphorylation of glycogen synthase kinase 3β in pancreatic β-cells. J Biol Chem 2013; 288:24825-33. [PMID: 23853095 DOI: 10.1074/jbc.m113.495945] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In pancreatic β-cells, glucose induces the binding of the transcription factor pancreatic duodenal homeobox-1 (PDX-1) to the insulin gene promoter to activate insulin gene transcription. At low glucose levels, glycogen synthase kinase 3β (GSK3β) is known to phosphorylate PDX-1 on C-terminal serine residues, which triggers PDX-1 proteasomal degradation. We previously showed that the serine/threonine Per-Arnt-Sim domain-containing kinase (PASK) regulates insulin gene transcription via PDX-1. However, the mechanisms underlying this regulation are unknown. In this study, we aimed to identify the role of PASK in the regulation of PDX-1 phosphorylation, protein expression, and stability in insulin-secreting cells and isolated rodent islets of Langerhans. We observed that glucose induces a decrease in overall PDX-1 serine phosphorylation and that overexpression of WT PASK mimics this effect. In vitro, PASK directly phosphorylates GSK3β on its inactivating phosphorylation site Ser(9). Overexpression of a kinase-dead (KD), dominant negative version of PASK blocks glucose-induced Ser(9) phosphorylation of GSK3β. Accordingly, GSK3β Ser(9) phosphorylation is reduced in islets from pask-null mice. Overexpression of WT PASK or KD GSK3β protects PDX-1 from degradation and results in increased PDX-1 protein abundance. Conversely, overexpression of KD PASK blocks glucose-induction of PDX-1 protein. We conclude that PASK phosphorylates and inactivates GSK3β, thereby preventing PDX-1 serine phosphorylation and alleviating GSK3β-mediated PDX-1 protein degradation in pancreatic β-cells.
Collapse
Affiliation(s)
- Meriem Semache
- Montreal Diabetes Research Center, CRCHUM, Quebec City H1W4A4, Canada
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Somesh BP, Verma MK, Sadasivuni MK, Mammen-Oommen A, Biswas S, Shilpa PC, Reddy AK, Yateesh AN, Pallavi PM, Nethra S, Smitha R, Neelima K, Narayanan U, Jagannath MR. Chronic glucolipotoxic conditions in pancreatic islets impair insulin secretion due to dysregulated calcium dynamics, glucose responsiveness and mitochondrial activity. BMC Cell Biol 2013; 14:31. [PMID: 23815372 PMCID: PMC3704974 DOI: 10.1186/1471-2121-14-31] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 06/24/2013] [Indexed: 11/10/2022] Open
Abstract
Background In the progression towards diabetes, glucolipotoxicity is one of the main causes of pancreatic beta cell pathology. The aim of this study was to examine the in vitro effects of chronic glucolipotoxic conditions on cellular responses in pancreatic islets, including glucose and fat metabolism, Calcium mobilization, insulin secretion and insulin content. Results Exposure of islets to chronic glucolipotoxic conditions decreased glucose stimulated insulin secretion in vitro. Reduced protein levels of Glut2/slc2a2, and decreased glucokinase and pyruvate carboxylase mRNA levels indicated a significant lowering in glucose sensing. Concomitantly, both fatty acid uptake and triglyceride accumulation increased significantly while fatty acid oxidation decreased. This general suppression in glucose metabolism correlated well with a decrease in mitochondrial number and activity, reduction in cellular ATP content and dampening of the TCA cycle. Further, we also observed a decrease in IP3 levels and lower Calcium mobilization in response to glucose. Importantly, chronic glucolipotoxic conditions in vitro decreased insulin gene expression, insulin content, insulin granule docking (to the plasma membrane) and insulin secretion. Conclusions Our results present an integrated view of the effects of chronic glucolipotoxic conditions on known and novel signaling events, in vitro, that results in reduced glucose responsiveness and insulin secretion.
Collapse
Affiliation(s)
- Baggavalli P Somesh
- Connexios Life Sciences Pvt Ltd., No. 49, First Main road, 3rd phase, JP Nagar, Bangalore 560 078, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Lamontagne J, Jalbert-Arsenault É, Pepin É, Peyot ML, Ruderman NB, Nolan CJ, Joly E, Madiraju SM, Poitout V, Prentki M. Pioglitazone acutely reduces energy metabolism and insulin secretion in rats. Diabetes 2013; 62:2122-9. [PMID: 23378607 PMCID: PMC3661607 DOI: 10.2337/db12-0428] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Our objective was to determine if the insulin-sensitizing drug pioglitazone acutely reduces insulin secretion and causes metabolic deceleration in vivo independently of change in insulin sensitivity. We assessed glucose homeostasis by hyperinsulinemic-euglycemic and hyperglycemic clamp studies and energy expenditure by indirect calorimetry and biotelemetry in male Wistar and obese hyperinsulinemic Zucker diabetic fatty (ZDF) rats 45 min after a single oral dose of pioglitazone (30 mg/kg). In vivo insulin secretion during clamped hyperglycemia was reduced in both Wistar and ZDF rats after pioglitazone administration. Insulin clearance was slightly increased in Wistar but not in ZDF rats. Insulin sensitivity in Wistar rats assessed by the hyperinsulinemic-euglycemic clamp was minimally affected by pioglitazone at this early time point. Pioglitazone also reduced energy expenditure in Wistar rats without altering respiratory exchange ratio or core body temperature. Glucose-induced insulin secretion (GIIS) and oxygen consumption were reduced by pioglitazone in isolated islets and INS832/13 cells. In conclusion, pioglitazone acutely induces whole-body metabolic slowing down and reduces GIIS, the latter being largely independent of the insulin-sensitizing action of the drug. The results suggest that pioglitazone has direct metabolic deceleration effects on the β-cell that may contribute to its capacity to lower insulinemia and antidiabetic action.
Collapse
Affiliation(s)
- Julien Lamontagne
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
- Department of Biochemistry, Université de Montréal, Montreal, Quebec, Canada
| | - Élise Jalbert-Arsenault
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Émilie Pepin
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
- Program of Molecular Biology, Université de Montréal, Montreal, Quebec, Canada
| | - Marie-Line Peyot
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
| | - Neil B. Ruderman
- Departments of Medicine and Physiology and Biophysics, Boston University School of Medicine and Diabetes Unit, Section of Endocrinology, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | | | - Erik Joly
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
| | - S.R. Murthy Madiraju
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center at the CRCHUM and Departments of Medicine, Biochemistry and Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Marc Prentki
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
- Department of Biochemistry, Université de Montréal, Montreal, Quebec, Canada
- Corresponding author: Marc Prentki,
| |
Collapse
|
32
|
Green-Mitchell SM, Tersey SA, Cole BK, Ma K, Kuhn NS, Cunningham TD, Maybee NA, Chakrabarti SK, McDuffie M, Taylor-Fishwick DA, Mirmira RG, Nadler JL, Morris MA. Deletion of 12/15-lipoxygenase alters macrophage and islet function in NOD-Alox15(null) mice, leading to protection against type 1 diabetes development. PLoS One 2013; 8:e56763. [PMID: 23437231 PMCID: PMC3578926 DOI: 10.1371/journal.pone.0056763] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 01/16/2013] [Indexed: 11/19/2022] Open
Abstract
AIMS Type 1 diabetes (T1D) is characterized by autoimmune depletion of insulin-producing pancreatic beta cells. We showed previously that deletion of the 12/15-lipoxygenase enzyme (12/15-LO, Alox15 gene) in NOD mice leads to nearly 100 percent protection from T1D. In this study, we test the hypothesis that cytokines involved in the IL-12/12/15-LO axis affect both macrophage and islet function, which contributes to the development of T1D. METHODS 12/15-LO expression was clarified in immune cells by qRT-PCR, and timing of expression was tested in islets using qRT-PCR and Western blotting. Expression of key proinflammatory cytokines and pancreatic transcription factors was studied in NOD and NOD-Alox15(null) macrophages and islets using qRT-PCR. The two mouse strains were also assessed for the ability of splenocytes to transfer diabetes in an adoptive transfer model, and beta cell mass. RESULTS 12/15-LO is expressed in macrophages, but not B and T cells of NOD mice. In macrophages, 12/15-LO deletion leads to decreased proinflammatory cytokine mRNA and protein levels. Furthermore, splenocytes from NOD-Alox15(null) mice are unable to transfer diabetes in an adoptive transfer model. In islets, expression of 12/15-LO in NOD mice peaks at a crucial time during insulitis development. The absence of 12/15-LO results in maintenance of islet health with respect to measurements of islet-specific transcription factors, markers of islet health, proinflammatory cytokines, and beta cell mass. CONCLUSIONS These results suggest that 12/15-LO affects islet and macrophage function, causing inflammation, and leading to autoimmunity and reduced beta cell mass.
Collapse
Affiliation(s)
- Shamina M. Green-Mitchell
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Sarah A. Tersey
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Banumathi K. Cole
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Kaiwen Ma
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Norine S. Kuhn
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Tina Duong Cunningham
- Graduate Program in Public Health, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Nelly A. Maybee
- Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Swarup K. Chakrabarti
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Marcia McDuffie
- Department of Microbiology, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - David A. Taylor-Fishwick
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Raghavendra G. Mirmira
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Jerry L. Nadler
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
- Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Margaret A. Morris
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| |
Collapse
|
33
|
Zou R, Yang L, Xue J, Ke M, Huang Q, Huang Q, Dai Z, Sun J, Xu Y. WITHDRAWN: RIP140 mediates hyperglycemia-induced glucotoxicity in β-cells via the activation of JNK and ERK1/2 signaling pathways. Diabetes Res Clin Pract 2013:S0168-8227(12)00506-2. [PMID: 23290273 DOI: 10.1016/j.diabres.2012.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 10/22/2012] [Accepted: 12/13/2012] [Indexed: 10/27/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Runmei Zou
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Donghu Road 169#, Wuhan, Hubei 430071, China
| | - Li Yang
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Donghu Road 169#, Wuhan, Hubei 430071, China
| | - Junli Xue
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Donghu Road 169#, Wuhan, Hubei 430071, China
| | - Min Ke
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Donghu Road 169#, Wuhan, Hubei 430071, China
| | - Qian Huang
- Department of Paediatrics, Zhongnan Hospital of Wuhan University, Donghu Road 169#, Wuhan, Hubei 430071, China
| | - Qi Huang
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Donghu Road 169#, Wuhan, Hubei 430071, China
| | - Zhe Dai
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Donghu Road 169#, Wuhan, Hubei 430071, China
| | - Jiazhong Sun
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Donghu Road 169#, Wuhan, Hubei 430071, China
| | - Yancheng Xu
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Donghu Road 169#, Wuhan, Hubei 430071, China.
| |
Collapse
|
34
|
Maris M, Robert S, Waelkens E, Derua R, Hernangomez MH, D'Hertog W, Cnop M, Mathieu C, Overbergh L. Role of the saturated nonesterified fatty acid palmitate in beta cell dysfunction. J Proteome Res 2012; 12:347-62. [PMID: 23170928 DOI: 10.1021/pr300596g] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sustained elevated levels of saturated free fatty acids, such as palmitate, contribute to beta cell dysfunction, a phenomenon aggravated by high glucose levels. The aim of this study was to investigate the mechanisms of palmitate-induced beta cell dysfunction and death, combined or not with high glucose. Protein profiling of INS-1E cells, exposed to 0.5 mmol/L palmitate and combined or not with 25 mmol/L glucose, for 24 h was done by 2D-DIGE, both on full cell lysate and on an enriched endoplasmic reticulum (ER) fraction. Eighty-three differentially expressed proteins (P < 0.05) were identified by MALDI-TOF/TOF mass spectrometry and proteomic results were confirmed by functional assays. 2D-DIGE analysis of whole cell lysates and ER enriched samples revealed a high number of proteins compared to previous reports. Palmitate induced beta cell dysfunction and death via ER stress, hampered insulin maturation, generation of harmful metabolites during triglycerides synthesis and altered intracellular trafficking. In combination with high glucose, palmitate induced increased shunting of excess glucose, increased mitochondrial reactive oxygen species production and an elevation in many transcription-related proteins. This study contributes to a better understanding and revealed novel mechanisms of palmitate-induced beta cell dysfunction and death and may provide new targets for drug discovery.
Collapse
Affiliation(s)
- Michael Maris
- Laboratory of Clinical and Experimental Endocrinology, Herestraat 49, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Gosmanov AR, Smiley DD, Peng L, Siquiera J, Robalino G, Newton C, Umpierrez GE. Vascular effects of intravenous intralipid and dextrose infusions in obese subjects. Metabolism 2012; 61:1370-6. [PMID: 22483976 PMCID: PMC3738183 DOI: 10.1016/j.metabol.2012.03.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Revised: 03/08/2012] [Accepted: 03/09/2012] [Indexed: 01/22/2023]
Abstract
Hyperglycemia and elevated free fatty acids (FFA) are implicated in the development of endothelial dysfunction. Infusion of soy-bean oil-based lipid emulsion (Intralipid®) increases FFA levels and results in elevation of blood pressure (BP) and endothelial dysfunction in obese healthy subjects. The effects of combined hyperglycemia and high FFA on BP, endothelial function and carbohydrate metabolism are not known. Twelve obese healthy subjects received four random, 8-h IV infusions of saline, Intralipid 40 mL/h, Dextrose 10% 40 mL/h, or combined Intralipid and dextrose. Plasma levels of FFA increased by 1.03±0.34 mmol/L (p=0.009) after Intralipid, but FFAs remained unchanged during saline, dextrose, and combined Intralipid and dextrose infusion. Plasma glucose and insulin concentrations significantly increased after dextrose and combined Intralipid and dextrose (all, p<0.05) and were not different from baseline during saline and lipid infusion. Intralipid increased systolic BP by 12±9 mmHg (p<0.001) and diastolic BP by 5±6 mmHg (p=0.022),and decreased flow-mediated dilatation (FMD) from baseline by 3.2%±1.4% (p<0.001). Saline and dextrose infusion had neutral effects on BP and FMD. The co-administration of lipid and dextrose decreased FMD by 2.4%±2.1% (p=0.002) from baseline, but did not significantly increase systolic or diastolic BP. Short-term Intralipid infusion significantly increased FFA and BP; in contrast, FFA and BP were unchanged during combined infusion of Intralipid and dextrose. Combined Intralipid and dextrose infusion resulted in endothelial dysfunction similar to Intralipid alone.
Collapse
Affiliation(s)
- Aidar R. Gosmanov
- Department of Medicine, Division of Endocrinology University of Tennessee Health Science Center, Memphis, TN
| | - Dawn D. Smiley
- Department of Medicine, Division of Endocrinology, Emory University, Atlanta, GA
| | - Limin Peng
- Rollins School of Public Health, Emory University, Atlanta, GA
| | - Joselita Siquiera
- Department of Medicine, Division of Endocrinology, Emory University, Atlanta, GA
| | - Gonzalo Robalino
- Department of Medicine, Division of Endocrinology, Emory University, Atlanta, GA
| | - Christopher Newton
- Department of Medicine, Division of Endocrinology, Emory University, Atlanta, GA
| | - Guillermo E. Umpierrez
- Department of Medicine, Division of Endocrinology, Emory University, Atlanta, GA
- Corresponding author. Emory University School of Medicine, 49 Jesse Hill Jr. Drive, Atlanta, Georgia 30303. Tel.: +1 404 778 1665; fax: +1 404 778 1661. (G.E. Umpierrez)
| |
Collapse
|
36
|
Salgin B, Ong KK, Thankamony A, Emmett P, Wareham NJ, Dunger DB. Higher fasting plasma free fatty acid levels are associated with lower insulin secretion in children and adults and a higher incidence of type 2 diabetes. J Clin Endocrinol Metab 2012; 97:3302-9. [PMID: 22740706 DOI: 10.1210/jc.2012-1428] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
CONTEXT There are limited data in humans on the association between fasting free fatty acid (FFA) levels and pancreatic β-cell function. OBJECTIVE Our objective was to examine this association in children and adults with normal glucose tolerance and to explore fasting FFA levels in relation to subsequent risk of impaired glucose tolerance (IGT) and type 2 diabetes (T2D). DESIGN We measured FFA, glucose, and insulin levels after an overnight fast and 30 min after an oral glucose load in 797 children aged 8 yr in the Avon Longitudinal Study of Parents and Children and 770 adults aged 44-71 yr in the Medical Research Council Ely Study. We calculated the homeostasis model assessment to estimate fasting insulin sensitivity, the insulinogenic index to estimate insulin secretion, and the disposition index to assess insulin secretion corrected for insulin sensitivity. RESULTS Higher fasting FFA levels were associated with lower insulin secretion in children (boys, P = 0.03; girls, P = 0.001) and adults (men, P = 0.03, women, P = 0.04). Associations with insulin sensitivity were more variable, but after adjustment for insulin sensitivity, higher fasting FFA levels remained associated with lower insulin secretion (disposition index). Compared with adults in the lowest tertile of fasting FFA levels, those in the middle and highest tertiles had a 3-fold higher incidence of IGT or T2D over the following 5-8 yr. CONCLUSIONS Higher fasting FFA levels were consistently associated with lower insulin secretion in children and adults with normal glucose tolerance. Furthermore, higher fasting FFA levels were prospectively associated with a greater risk of subsequent IGT and T2D.
Collapse
Affiliation(s)
- Burak Salgin
- University Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, United Kingdom.
| | | | | | | | | | | |
Collapse
|
37
|
Activation of SIRT1 protects pancreatic β-cells against palmitate-induced dysfunction. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1815-25. [PMID: 22968147 DOI: 10.1016/j.bbadis.2012.08.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 08/08/2012] [Accepted: 08/13/2012] [Indexed: 01/23/2023]
Abstract
Sirtuin 1 (SIRT1), a nicotinamide adenosine dinucleotide-dependent histone deacetylase, is an important regulator of energy homeostasis in response to nutrient availability. In pancreatic β-cells, SIRT1 has been shown to up-regulate insulin secretion in response to glucose stimulation. However, the potential roles of SIRT1 in islet β-cells against lipotoxicity remain poorly understood. Here, we demonstrated that SIRT1 mRNA and protein expressions were markedly reduced in the islets isolated from rats infused with 20% Intralipid for 24h. Long-term exposure to 0.4mmol/L palmitate also decreased SIRT1 expression in cultured INS-1 cells and isolated rat islets, which was prevented by 10μmol/L resveratrol, a SIRT1 agonist. In addition, resveratrol improved glucose-stimulated insulin secretion decreased by palmitate, which was abrogated by EX527, a specific SIRT1 inhibitor. Furthermore, inhibition of SIRT1 activity by EX527 or a knockdown of SIRT1 suppressed insulin promoter activity, along with decreased insulin, v-maf musculoaponeurotic fibrosarcoma oncogene homolog A (MafA), and NK6 homeodomain 1 (NKX6.1) mRNA expressions. Activation of SIRT1 by resveratrol or overexpression of SIRT1 antagonized palmitate-inhibited insulin transcriptional activity. SIRT1 overexpression exerted an additive effect on pancreatic and duodenal homeobox 1 (PDX1)-stimulated insulin promoter activity and abolished forkhead box O1 protein (FOXO1)-decreased insulin transcriptional activity. Resveratrol reversed FOXO1 nuclear translocation induced by palmitate. Our findings indicate that SIRT1 protects against palmitate-induced β-cell dysfunction.
Collapse
|
38
|
|
39
|
Lipopolysaccharides impair insulin gene expression in isolated islets of Langerhans via Toll-Like Receptor-4 and NF-κB signalling. PLoS One 2012; 7:e36200. [PMID: 22558381 PMCID: PMC3338606 DOI: 10.1371/journal.pone.0036200] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 04/03/2012] [Indexed: 12/12/2022] Open
Abstract
Background Type 2 diabetes is characterized by pancreatic β-cell dysfunction and is associated with low-grade inflammation. Recent observations suggest that the signalling cascade activated by lipopolysaccharides (LPS) binding to Toll-Like Receptor 4 (TLR4) exerts deleterious effects on pancreatic β-cell function; however, the molecular mechanisms of these effects are incompletely understood. In this study, we tested the hypothesis that LPS alters insulin gene expression via TLR4 and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in islets. Methodology/Principal Findings A 24-h exposure of isolated human, rat and mouse islets of Langerhans to LPS dose-dependently reduced insulin gene expression. This was associated in mouse and rat islets with decreased mRNA expression of pancreas-duodenum homebox-1 (PDX-1) and mammalian homologue of avian MafA/l-Maf (MafA). Accordingly, LPS exposure also decreased glucose-induced insulin secretion. LPS repression of insulin, PDX-1 and MafA expression, as well as its inhibition of insulin secretion, were not observed in islets from TLR4-deficient mice. LPS inhibition of β-cell gene expression in rat islets was prevented by inhibition of the NF-κB pathway, but not the p38 mitogen-activated protein kinase (p38 MAPK) pathway. Conclusions/Significance Our findings demonstrate that LPS inhibit β-cell gene expression in a TLR4-dependent manner and via NF-κB signaling in pancreatic islets, suggesting a novel mechanism by which the gut microbiota might affect pancreatic β-cell function.
Collapse
|
40
|
Pascoe J, Hollern D, Stamateris R, Abbasi M, Romano LC, Zou B, O’Donnell CP, Garcia-Ocana A, Alonso LC. Free fatty acids block glucose-induced β-cell proliferation in mice by inducing cell cycle inhibitors p16 and p18. Diabetes 2012; 61:632-41. [PMID: 22338094 PMCID: PMC3282818 DOI: 10.2337/db11-0991] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Pancreatic β-cell proliferation is infrequent in adult humans and is not increased in type 2 diabetes despite obesity and insulin resistance, suggesting the existence of inhibitory factors. Free fatty acids (FFAs) may influence proliferation. In order to test whether FFAs restrict β-cell proliferation in vivo, mice were intravenously infused with saline, Liposyn II, glucose, or both, continuously for 4 days. Lipid infusion did not alter basal β-cell proliferation, but blocked glucose-stimulated proliferation, without inducing excess β-cell death. In vitro exposure to FFAs inhibited proliferation in both primary mouse β-cells and in rat insulinoma (INS-1) cells, indicating a direct effect on β-cells. Two of the fatty acids present in Liposyn II, linoleic acid and palmitic acid, both reduced proliferation. FFAs did not interfere with cyclin D2 induction or nuclear localization by glucose, but increased expression of inhibitor of cyclin dependent kinase 4 (INK4) family cell cycle inhibitors p16 and p18. Knockdown of either p16 or p18 rescued the antiproliferative effect of FFAs. These data provide evidence for a novel antiproliferative form of β-cell glucolipotoxicity: FFAs restrain glucose-stimulated β-cell proliferation in vivo and in vitro through cell cycle inhibitors p16 and p18. If FFAs reduce proliferation induced by obesity and insulin resistance, targeting this pathway may lead to new treatment approaches to prevent diabetes.
Collapse
Affiliation(s)
- Jordan Pascoe
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Douglas Hollern
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rachel Stamateris
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Munira Abbasi
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lia C. Romano
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Baobo Zou
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christopher P. O’Donnell
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adolfo Garcia-Ocana
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Laura C. Alonso
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
- Corresponding author: Laura C. Alonso,
| |
Collapse
|
41
|
Han SJ, Choi SE, Yi SA, Lee SJ, Kim HJ, Kim DJ, Lee HC, Lee KW, Kang Y. β-Cell-protective effect of 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid as a glutamate dehydrogenase activator in db/db mice. J Endocrinol 2012; 212:307-15. [PMID: 22131441 DOI: 10.1530/joe-11-0340] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
2-Aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (BCH) is an activator of glutamate dehydrogenase (GDH), which is a mitochondrial enzyme with an important role in insulin secretion. We investigated the effect of BCH on the high-glucose (HG)-induced reduction in glucose-stimulated insulin secretion (GSIS), the HG/palmitate (PA)-induced reduction in insulin gene expression, and HG/PA-induced β-cell death. We also studied whether long-term treatment with BCH lowers blood glucose and improves β-cell integrity in db/db mice. We evaluated GSIS, insulin gene expression, and DNA fragmentation in INS-1 cells exposed to HG or HG/PA in the presence or absence of BCH. An in vivo study was performed in which 7-week-old diabetic db/db mice were treated with BCH (0.7 g/kg, n = 10) and placebo (n = 10) every other day for 6 weeks. After treatment, an intraperitoneal glucose tolerance test and immunohistological examinations were performed. Treatment with BCH blocked HG-induced GSIS inhibition and the HG/PA-induced reduction in insulin gene expression in INS-1 cells. In addition, BCH significantly reduced HG/PA-induced INS-1 cell death and phospho-JNK level. BCH treatment improved glucose tolerance and insulin secretion in db/db mice. BCH treatment also increased the ratio of insulin-positive β-cells to total islet area (P < 0.05) and reduced the percentage of β-cells expressing cleaved caspase 3 (P < 0.05). In conclusion, the GDH activator BCH improved glycemic control in db/db mice. This anti-diabetic effect may be associated with improved insulin secretion, preserved islet architecture, and reduced β-cell apoptosis.
Collapse
Affiliation(s)
- Seung Jin Han
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, San 5, Wonchon-dong, Yeongtong-gu, Suwon 443-721, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mediation of glucolipotoxicity in INS-1 rat insulinoma cells by small heterodimer partner interacting leucine zipper protein (SMILE). Biochem Biophys Res Commun 2012; 419:768-73. [PMID: 22387546 DOI: 10.1016/j.bbrc.2012.02.098] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 02/16/2012] [Indexed: 12/25/2022]
Abstract
Sustained elevations of glucose and free fatty acid concentration have deleterious effects on pancreatic beta cell function. One of the hallmarks of such glucolipotoxicity is a reduction in insulin gene expression, resulting from decreased insulin promoter activity. Sterol regulatory element binding protein-1c (SREBP-1c), a lipogenic transcription factor, is related to the development of beta cell dysfunction caused by elevated concentrations of glucose and free fatty acid. Small heterodimer partner (SHP) interacting leucine zipper protein (SMILE), also known as Zhangfei, is a novel protein which interacts with SHP that mediates glucotoxicity in INS-1 rat insulinoma cells. Treatment of INS-1 cells with high concentrations of glucose and palmitate increased SREBP-1c and SMILE expression, and decreased insulin gene expression. Adenovirus-mediated overexpression of SREBP-1c in INS-1 cells induced SMILE expression. Moreover, adenovirus-mediated overexpression of SMILE (Ad-SMILE) in INS-1 cells impaired glucose-stimulated insulin secretion as well as insulin gene expression. Ad-SMILE overexpression also inhibited the expression of beta-cell enriched transcription factors including pancreatic duodenal homeobox factor-1, beta cell E box transactivator 2 and RIPE3b1/MafA, in INS-1 cells. Finally, in COS-1 cells, expression of SMILE inhibited the insulin promoter activity induced by these same beta-cell enriched transcription factors. These results collectively suggest that SMILE plays an important role in the development of beta cell dysfunction induced by glucolipotoxicity.
Collapse
|
43
|
Amyot J, Benterki I, Fontés G, Hagman DK, Ferdaoussi M, Teodoro T, Volchuk A, Joly É, Poitout V. Binding of activating transcription factor 6 to the A5/Core of the rat insulin II gene promoter does not mediate its transcriptional repression. J Mol Endocrinol 2011; 47:273-83. [PMID: 21821716 PMCID: PMC3185209 DOI: 10.1530/jme-11-0016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic β-cells have a well-developed endoplasmic reticulum due to their highly specialized secretory function to produce insulin in response to glucose and nutrients. It has been previously reported that overexpression of activating transcription factor 6 (ATF6) reduces insulin gene expression in part via upregulation of small heterodimer partner. In this study, we investigated whether ATF6 directly binds to the insulin gene promoter, and whether its direct binding represses insulin gene promoter activity. A bioinformatics analysis identified a putative ATF6 binding site in the A5/Core region of the rat insulin II gene promoter. Direct binding of ATF6 was confirmed using several approaches. Electrophoretic mobility shift assays in nuclear extracts from MCF7 cells, isolated rat islets and insulin-secreting HIT-T15 cells showed ATF6 binding to the native A5/Core of the rat insulin II gene promoter. Antibody-mediated supershift analyses revealed the presence of both ATF6 isoforms, ATF6α and ATF6β, in the complex. Chromatin immunoprecipitation assays confirmed the binding of ATF6α and ATF6β to a region encompassing the A5/Core of the rat insulin II gene promoter in isolated rat islets. Overexpression of the active (cleaved) fragment of ATF6α, but not ATF6β, inhibited the activity of an insulin promoter-reporter by 50%. However, the inhibitory effect of ATF6α was insensitive to mutational inactivation or deletion of the A5/Core. Therefore, although ATF6 binds directly to the A5/Core of the rat insulin II gene promoter, this direct binding does not appear to contribute to its repressive activity.
Collapse
Affiliation(s)
- Julie Amyot
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Technopole Angus, 2901 Rachel Est, Montréal, QC, Canada, H1W 4A4
- Department of Biochemistry, Université de Montréal, C.P. 6128, succursale Centre-ville, QC, Canada, H3C 3J7
| | - Isma Benterki
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Technopole Angus, 2901 Rachel Est, Montréal, QC, Canada, H1W 4A4
- Department of Biochemistry, Université de Montréal, C.P. 6128, succursale Centre-ville, QC, Canada, H3C 3J7
| | - Ghislaine Fontés
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Technopole Angus, 2901 Rachel Est, Montréal, QC, Canada, H1W 4A4
| | - Derek K. Hagman
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Technopole Angus, 2901 Rachel Est, Montréal, QC, Canada, H1W 4A4
| | - Mourad Ferdaoussi
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Technopole Angus, 2901 Rachel Est, Montréal, QC, Canada, H1W 4A4
| | - Tracy Teodoro
- Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, 101 College Street, TMDT 10-706, Toronto, ON, Canada, M5G1L7
| | - Allen Volchuk
- Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, 101 College Street, TMDT 10-706, Toronto, ON, Canada, M5G1L7
| | - Érik Joly
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Technopole Angus, 2901 Rachel Est, Montréal, QC, Canada, H1W 4A4
| | - Vincent Poitout
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Technopole Angus, 2901 Rachel Est, Montréal, QC, Canada, H1W 4A4
- Department of Biochemistry, Université de Montréal, C.P. 6128, succursale Centre-ville, QC, Canada, H3C 3J7
- Department of Medicine, Université de Montréal, C.P. 6128, succursale Centre-ville, QC, Canada, H3C 3J7
- Department of Nutrition, Université de Montréal, C.P. 6128, succursale Centre-ville, QC, Canada, H3C 3J7
| |
Collapse
|
44
|
Giacca A, Xiao C, Oprescu AI, Carpentier AC, Lewis GF. Lipid-induced pancreatic β-cell dysfunction: focus on in vivo studies. Am J Physiol Endocrinol Metab 2011; 300:E255-62. [PMID: 21119027 DOI: 10.1152/ajpendo.00416.2010] [Citation(s) in RCA: 157] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The phenomenon of lipid-induced pancreatic β-cell dysfunction ("lipotoxicity") has been very well documented in numerous in vitro experimental systems and has become widely accepted. In vivo demonstration of β-cell lipotoxicity, on the other hand, has not been consistently demonstrated, and there remains a lack of consensus regarding the in vivo effects of chronically elevated free fatty acids (FFA) on β-cell function. Much of the disagreement relates to how insulin secretion is quantified in vivo and in particular whether insulin secretion is assessed in relation to whole body insulin sensitivity, which is clearly reduced by elevated FFA. By correcting for changes in in vivo insulin sensitivity, we and others have shown that prolonged elevation of FFA impairs β-cell secretory function. Prediabetic animal models and humans with a positive family history of type 2 diabetes are more susceptible to this impairment, whereas those with severe impairment of β-cell function (such as individuals with type 2 diabetes) demonstrate no additional impairment of β-cell function when FFA are experimentally raised. Glucolipotoxicity (i.e., the combined β-cell toxicity of elevated glucose and FFA) has been amply demonstrated in vitro and in some animal studies but not in humans, perhaps because there are limitations in experimentally raising plasma glucose to sufficiently high levels for prolonged periods of time. We and others have shown that therapies directed toward diminishing oxidative stress and ER stress have the potential to reduce lipid-induced β-cell dysfunction in animals and humans. In conclusion, lipid-induced pancreatic β-cell dysfunction is likely to be one contributor to the complex array of genetic and metabolic insults that result in the relentless decline in pancreatic β-cell function in those destined to develop type 2 diabetes, and mechanisms involved in this lipotoxicity are promising therapeutic targets.
Collapse
Affiliation(s)
- Adria Giacca
- Dept. of Physiology, Univ. of Toronto, ON, Canada.
| | | | | | | | | |
Collapse
|
45
|
Maris M, Ferreira GB, D’Hertog W, Cnop M, Waelkens E, Overbergh L, Mathieu C. High Glucose Induces Dysfunction in Insulin Secretory Cells by Different Pathways: A Proteomic Approach. J Proteome Res 2010; 9:6274-87. [DOI: 10.1021/pr100557w] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Michael Maris
- Laboratory for Experimental Medicine and Endocrinology (LEGENDO), Herestraat 49, Catholic University of Leuven, Leuven, Belgium, Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070 Brussels, Belgium, Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 1070 Brussels, Belgium, Laboratory of Protein Phosphorylation and Proteomics, Catholic University of Leuven, Leuven, Belgium, and ProMeta, Catholic University of
| | - Gabriela B. Ferreira
- Laboratory for Experimental Medicine and Endocrinology (LEGENDO), Herestraat 49, Catholic University of Leuven, Leuven, Belgium, Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070 Brussels, Belgium, Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 1070 Brussels, Belgium, Laboratory of Protein Phosphorylation and Proteomics, Catholic University of Leuven, Leuven, Belgium, and ProMeta, Catholic University of
| | - Wannes D’Hertog
- Laboratory for Experimental Medicine and Endocrinology (LEGENDO), Herestraat 49, Catholic University of Leuven, Leuven, Belgium, Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070 Brussels, Belgium, Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 1070 Brussels, Belgium, Laboratory of Protein Phosphorylation and Proteomics, Catholic University of Leuven, Leuven, Belgium, and ProMeta, Catholic University of
| | - Miriam Cnop
- Laboratory for Experimental Medicine and Endocrinology (LEGENDO), Herestraat 49, Catholic University of Leuven, Leuven, Belgium, Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070 Brussels, Belgium, Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 1070 Brussels, Belgium, Laboratory of Protein Phosphorylation and Proteomics, Catholic University of Leuven, Leuven, Belgium, and ProMeta, Catholic University of
| | - Etienne Waelkens
- Laboratory for Experimental Medicine and Endocrinology (LEGENDO), Herestraat 49, Catholic University of Leuven, Leuven, Belgium, Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070 Brussels, Belgium, Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 1070 Brussels, Belgium, Laboratory of Protein Phosphorylation and Proteomics, Catholic University of Leuven, Leuven, Belgium, and ProMeta, Catholic University of
| | - Lut Overbergh
- Laboratory for Experimental Medicine and Endocrinology (LEGENDO), Herestraat 49, Catholic University of Leuven, Leuven, Belgium, Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070 Brussels, Belgium, Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 1070 Brussels, Belgium, Laboratory of Protein Phosphorylation and Proteomics, Catholic University of Leuven, Leuven, Belgium, and ProMeta, Catholic University of
| | - Chantal Mathieu
- Laboratory for Experimental Medicine and Endocrinology (LEGENDO), Herestraat 49, Catholic University of Leuven, Leuven, Belgium, Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070 Brussels, Belgium, Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 1070 Brussels, Belgium, Laboratory of Protein Phosphorylation and Proteomics, Catholic University of Leuven, Leuven, Belgium, and ProMeta, Catholic University of
| |
Collapse
|
46
|
Fontés G, Zarrouki B, Hagman DK, Latour MG, Semache M, Roskens V, Moore PC, Prentki M, Rhodes CJ, Jetton TL, Poitout V. Glucolipotoxicity age-dependently impairs beta cell function in rats despite a marked increase in beta cell mass. Diabetologia 2010; 53:2369-79. [PMID: 20628728 PMCID: PMC2947580 DOI: 10.1007/s00125-010-1850-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 06/18/2010] [Indexed: 01/25/2023]
Abstract
AIMS/HYPOTHESIS Prolonged exposure of pancreatic beta cells to excessive levels of glucose and fatty acids, referred to as glucolipotoxicity, is postulated to contribute to impaired glucose homeostasis in patients with type 2 diabetes. However, the relative contribution of defective beta cell function vs diminished beta cell mass under glucolipotoxic conditions in vivo remains a subject of debate. We therefore sought to determine whether glucolipotoxicity in rats is due to impaired beta cell function and/or reduced beta cell mass, and whether older animals are more susceptible to glucolipotoxic condition. METHODS Wistar rats (2 and 6 months old) received a 72 h infusion of glucose + intravenous fat emulsion or saline control. In vivo insulin secretion and sensitivity were assessed by hyperglycaemic clamps. Ex vivo insulin secretion, insulin biosynthesis and gene expression were measured in isolated islets. Beta cell mass and proliferation were examined by immunohistochemistry. RESULTS A 72 h infusion of glucose + intravenous fat emulsion in 2-month-old Wistar rats did not affect insulin sensitivity, insulin secretion or beta cell mass. In 6-month-old rats by contrast it led to insulin resistance and reduced insulin secretion in vivo, despite an increase in beta cell mass and proliferation. This was associated with: (1) diminished glucose-stimulated second-phase insulin secretion and proinsulin biosynthesis; (2) lower insulin content; and (3) reduced expression of beta cell genes in isolated islets. CONCLUSIONS/INTERPRETATION In this in vivo model, glucolipotoxicity is characterised by an age-dependent impairment of glucose-regulated beta cell function despite a marked increase in beta cell mass.
Collapse
Affiliation(s)
- G. Fontés
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada
- CRCHUM–Technopole Angus, 2901 Rachel Est, Montréal, QC, Canada H1W 4A4
- Department of Medicine, University of Montreal, Montreal, QC, Canada
| | - B. Zarrouki
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada
- CRCHUM–Technopole Angus, 2901 Rachel Est, Montréal, QC, Canada H1W 4A4
- Department of Medicine, University of Montreal, Montreal, QC, Canada
| | - D. K. Hagman
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada
- CRCHUM–Technopole Angus, 2901 Rachel Est, Montréal, QC, Canada H1W 4A4
- Department of Medicine, University of Montreal, Montreal, QC, Canada
| | - M. G. Latour
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada
- CRCHUM–Technopole Angus, 2901 Rachel Est, Montréal, QC, Canada H1W 4A4
| | - M. Semache
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada
- CRCHUM–Technopole Angus, 2901 Rachel Est, Montréal, QC, Canada H1W 4A4
- Department of Biochemistry, University of Montreal, Montreal, QC, Canada
| | - V. Roskens
- Division of Endocrinology, Diabetes and Metabolism, University of Vermont College of Medicine, Burlington, VT, USA
| | - P. C. Moore
- Kovler Diabetes Center, University of Chicago, Chicago, IL, USA
| | - M. Prentki
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada
- CRCHUM–Technopole Angus, 2901 Rachel Est, Montréal, QC, Canada H1W 4A4
- Department of Nutrition, University of Montreal, Montreal, QC, Canada
- Department of Biochemistry, University of Montreal, Montreal, QC, Canada
| | - C. J. Rhodes
- Kovler Diabetes Center, University of Chicago, Chicago, IL, USA
| | - T. L. Jetton
- Division of Endocrinology, Diabetes and Metabolism, University of Vermont College of Medicine, Burlington, VT, USA
| | - V. Poitout
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada
- CRCHUM–Technopole Angus, 2901 Rachel Est, Montréal, QC, Canada H1W 4A4,
- Department of Medicine, University of Montreal, Montreal, QC, Canada
- Department of Nutrition, University of Montreal, Montreal, QC, Canada
- Department of Biochemistry, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
47
|
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
|
48
|
Poitout V, Amyot J, Semache M, Zarrouki B, Hagman D, Fontés G. Glucolipotoxicity of the pancreatic beta cell. BIOCHIMICA ET BIOPHYSICA ACTA 2010; 1801:289-98. [PMID: 19715772 PMCID: PMC2824006 DOI: 10.1016/j.bbalip.2009.08.006] [Citation(s) in RCA: 280] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 08/13/2009] [Accepted: 08/13/2009] [Indexed: 02/07/2023]
Abstract
The concept of glucolipotoxicity refers to the combined, deleterious effects of elevated glucose and fatty acid levels on pancreatic beta-cell function and survival. Significant progress has been made in recent years towards a better understanding of the cellular and molecular basis of glucolipotoxicity in the beta cell. The permissive effect of elevated glucose on the detrimental actions of fatty acids stems from the influence of glucose on intracellular fatty acid metabolism, promoting the synthesis of cellular lipids. The combination of excessive levels of fatty acids and glucose therefore leads to decreased insulin secretion, impaired insulin gene expression, and beta-cell death by apoptosis, all of which probably have distinct underlying mechanisms. Recent studies from our laboratory have identified several pathways implicated in fatty acid inhibition of insulin gene expression, including the extracellular-regulated kinase (ERK1/2) pathway, the metabolic sensor Per-Arnt-Sim kinase (PASK), and the ATF6 branch of the unfolded protein response. We have also confirmed in vivo in rats that the decrease in insulin gene expression is an early defect which precedes any detectable abnormality in insulin secretion. While the role of glucolipotoxicity in humans is still debated, the inhibitory effects of chronically elevated fatty acid levels has been clearly demonstrated in several studies, at least in individuals genetically predisposed to developing type 2 diabetes. It is therefore likely that glucolipotoxicity contributes to beta-cell failure in type 2 diabetes as well as to the decline in beta-cell function observed after the onset of the disease.
Collapse
Affiliation(s)
- Vincent Poitout
- Montreal Diabetes Research Center, CRCHUM, Department of Medicine, University of Montreal, Montreal, QC, Canada.
| | | | | | | | | | | |
Collapse
|
49
|
Chen J, Fontes G, Saxena G, Poitout V, Shalev A. Lack of TXNIP protects against mitochondria-mediated apoptosis but not against fatty acid-induced ER stress-mediated beta-cell death. Diabetes 2010; 59:440-7. [PMID: 19875615 PMCID: PMC2809961 DOI: 10.2337/db09-0949] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE We have previously shown that lack of thioredoxin-interacting protein (TXNIP) protects against diabetes and glucotoxicity-induced beta-cell apoptosis. Because the role of TXNIP in lipotoxicity is unknown, the goal of the present study was to determine whether TXNIP expression is regulated by fatty acids and whether TXNIP deficiency also protects beta-cells against lipoapoptosis. RESARCH DESIGN AND METHODS: To determine the effects of fatty acids on beta-cell TXNIP expression, INS-1 cells and isolated islets were incubated with/without palmitate and rats underwent cyclic infusions of glucose and/or Intralipid prior to islet isolation and analysis by quantitative real-time RT-PCR and immunoblotting. Using primary wild-type and TXNIP-deficient islets, we then assessed the effects of palmitate on apoptosis (transferase-mediated dUTP nick-end labeling [TUNEL]), mitochondrial death pathway (cytochrome c release), and endoplasmic reticulum (ER) stress (binding protein [BiP], C/EBP homologous protein [CHOP]). Effects of TXNIP deficiency were also tested in the context of staurosporine (mitochondrial damage) or thapsigargin (ER stress). RESULTS Glucose elicited a dramatic increase in islet TXNIP expression both in vitro and in vivo, whereas fatty acids had no such effect and, when combined with glucose, even abolished the glucose effect. We also found that TXNIP deficiency does not effectively protect against palmitate or thapsigargin-induced beta-cell apoptosis, but specifically prevents staurosporine- or glucose-induced toxicity. CONCLUSIONS Our results demonstrate that unlike glucose, fatty acids do not induce beta-cell expression of proapoptotic TXNIP. They further reveal that TXNIP deficiency specifically inhibits the mitochondrial death pathway underlying beta-cell glucotoxicity, whereas it has very few protective effects against ER stress-mediated lipoapoptosis.
Collapse
Affiliation(s)
- Junqin Chen
- Department of Medicine, University of Wisconsin and William F. Middleton Veterans Administration Hospital, Madison, Wisconsin
| | - Ghislaine Fontes
- Montreal Diabetes Research Center, CRCHUM, and Department of Medicine, University of Montreal, Quebec, Canada
| | - Geetu Saxena
- Department of Medicine, University of Wisconsin and William F. Middleton Veterans Administration Hospital, Madison, Wisconsin
| | - Vincent Poitout
- Montreal Diabetes Research Center, CRCHUM, and Department of Medicine, University of Montreal, Quebec, Canada
| | - Anath Shalev
- Department of Medicine, University of Wisconsin and William F. Middleton Veterans Administration Hospital, Madison, Wisconsin
- Corresponding author: Anath Shalev,
| |
Collapse
|
50
|
Lack of preservation of insulin gene expression by a glucagon-like peptide 1 agonist or a dipeptidyl peptidase 4 inhibitor in an in vivo model of glucolipotoxicity. Diabetes Res Clin Pract 2010; 87:322-8. [PMID: 20092903 DOI: 10.1016/j.diabres.2009.12.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 12/10/2009] [Accepted: 12/15/2009] [Indexed: 10/19/2022]
Abstract
UNLABELLED Prolonged exposure of pancreatic beta-cells to elevated levels of glucose and fatty acids adversely affects insulin secretion and gene expression. AIM To examine whether the GLP-1 agonist exenatide or the inhibitor of the GLP-1-degrading enzyme dipeptidyl peptidase 4 (DPP-4) sitagliptin rescue insulin gene expression in rats infused for 72h with glucose+Intralipid, independently from their glucose-lowering action. METHODS Wistar rats were infused alternatively with glucose or Intralipid for cycles of 4h each for a total of 72h. The animals received exenatide (5microg/kg/day IV) or sitagliptin (5mg/kg/day IV) continuously starting 4 days prior to and continuing throughout the 3-day infusion period. RESULTS Plasma glucose, fatty acids, insulin and C-peptide levels were unaffected by exenatide or sitagliptin treatment during the infusion period. Insulin mRNA levels increased in response to the glucose infusion, but this increase was abolished in islets from rats receiving glucose+Intralipid. Neither exenatide nor sitagliptin administration rescued insulin mRNA in glucose+Intralipid infused rats. CONCLUSIONS Neither a GLP-1 agonist nor a DPP-4 inhibitor, at doses that do not alter blood glucose levels, prevented the inhibition of insulin gene expression in this in vivo model of glucolipotoxicity.
Collapse
|