1
|
Jang J, Kim Y, Song T, Park S, Kim HJ, Koh JH, Cho Y, Park SY, Sadayappan S, Kwak HB, Wolfe RR, Kim IY, Choi CS. Free essential amino acid feeding improves endurance during resistance training via DRP1-dependent mitochondrial remodelling. J Cachexia Sarcopenia Muscle 2024; 15:1651-1663. [PMID: 38881251 PMCID: PMC11446676 DOI: 10.1002/jcsm.13519] [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: 12/17/2023] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Loss of muscle strength and endurance with aging or in various conditions negatively affects quality of life. Resistance exercise training (RET) is the most powerful means to improve muscle mass and strength, but it does not generally lead to improvements in endurance capacity. Free essential amino acids (EAAs) act as precursors and stimuli for synthesis of both mitochondrial and myofibrillar proteins that could potentially confer endurance and strength gains. Thus, we hypothesized that daily consumption of a dietary supplement of nine free EAAs with RET improves endurance in addition to the strength gains by RET. METHODS Male C57BL6J mice (9 weeks old) were assigned to control (CON), EAA, RET (ladder climbing, 3 times a week), or combined treatment of EAA and RET (EAA + RET) groups. Physical functions focusing on strength or endurance were assessed before and after the interventions. Several analyses were performed to gain better insight into the mechanisms by which muscle function was improved. We determined cumulative rates of myofibrillar and mitochondrial protein synthesis using 2H2O labelling and mass spectrometry; assessed ex vivo contractile properties and in vitro mitochondrial function, evaluated neuromuscular junction (NMJ) stability, and assessed implicated molecular singling pathways. Furthermore, whole-body and muscle insulin sensitivity along with glucose metabolism, were evaluated using a hyperinsulinaemic-euglycaemic clamp. RESULTS EAA + RET increased muscle mass (10%, P < 0.05) and strength (6%, P < 0.05) more than RET alone, due to an enhanced rate of integrated muscle protein synthesis (19%, P < 0.05) with concomitant activation of Akt1/mTORC1 signalling. Muscle quality (muscle strength normalized to mass) was improved by RET (i.e., RET and EAA + RET) compared with sedentary groups (10%, P < 0.05), which was associated with increased AchR cluster size and MuSK activation (P < 0.05). EAA + RET also increased endurance capacity more than RET alone (26%, P < 0.05) by increasing both mitochondrial protein synthesis (53%, P < 0.05) and DRP1 activation (P < 0.05). Maximal respiratory capacity increased (P < 0.05) through activation of the mTORC1-DRP1 signalling axis. These favourable effects were accompanied by an improvement in basal glucose metabolism (i.e., blood glucose concentrations and endogenous glucose production vs. CON, P < 0.05). CONCLUSIONS Combined treatment with balanced free EAAs and RET may effectively promote endurance capacity as well as muscle strength through increased muscle protein synthesis, improved NMJ stability, and enhanced mitochondrial dynamics via mTORC1-DRP1 axis activation, ultimately leading to improved basal glucose metabolism.
Collapse
Affiliation(s)
- Jiwoong Jang
- Integrative Metabolic Fluxomics Lab, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Department of Internal Medicine, Gil Medical Center, Gachon University, Incheon, Korea
| | - Yeongmin Kim
- Integrative Metabolic Fluxomics Lab, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Korea
| | - Taejeong Song
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, Center for Cardiovascular Research, University of Cincinnati, Cincinnati, Ohio, USA
| | - Sanghee Park
- Integrative Metabolic Fluxomics Lab, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Korea
| | - Hee-Joo Kim
- Integrative Metabolic Fluxomics Lab, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Korea
| | - Jin-Ho Koh
- Integrative Metabolic Fluxomics Lab, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Korea
| | - Yoonil Cho
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Korea
| | - Shi-Young Park
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Gachon Biomedical Convergence Institute, Gachon University Gil Medical Center, Incheon, Korea
| | - Sakthivel Sadayappan
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, Center for Cardiovascular Research, University of Cincinnati, Cincinnati, Ohio, USA
| | - Hyo-Bum Kwak
- Department of Kinesiology, Inha University, Incheon, Korea
- Institute of Sports & Arts Convergence, Inha University, Incheon, Korea
- Department of Biomedical Science, Program in Biomedical Science & Engineering, Inha University, Incheon, Korea
| | - Robert R Wolfe
- Department of Geriatrics, Center for Translational Research in Aging and Longevity, Donald W. Reynolds Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Il-Young Kim
- Integrative Metabolic Fluxomics Lab, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Korea
| | - Cheol Soo Choi
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Department of Internal Medicine, Gil Medical Center, Gachon University, Incheon, Korea
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Korea
| |
Collapse
|
2
|
Sandforth L, Brachs S, Reinke J, Willmes D, Sancar G, Seigner J, Juarez-Lopez D, Sandforth A, McBride JD, Ma JX, Haufe S, Jordan J, Birkenfeld AL. Role of human Kallistatin in glucose and energy homeostasis in mice. Mol Metab 2024; 82:101905. [PMID: 38431218 PMCID: PMC10937158 DOI: 10.1016/j.molmet.2024.101905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/05/2024] Open
Abstract
OBJECTIVE Kallistatin (KST), also known as SERPIN A4, is a circulating, broadly acting human plasma protein with pleiotropic properties. Clinical studies in humans revealed reduced KST levels in obesity. The exact role of KST in glucose and energy homeostasis in the setting of insulin resistance and type 2 diabetes is currently unknown. METHODS Kallistatin mRNA expression in human subcutaneous white adipose tissue (sWAT) of 47 people with overweight to obesity of the clinical trial "Comparison of Low Fat and Low Carbohydrate Diets With Respect to Weight Loss and Metabolic Effects (B-SMART)" was measured. Moreover, we studied transgenic mice systemically overexpressing human KST (hKST-TG) and wild type littermate control mice (WT) under normal chow (NCD) and high-fat diet (HFD) conditions. RESULTS In sWAT of people with overweight to obesity, KST mRNA increased after diet-induced weight loss. On NCD, we did not observe differences between hKST-TG and WT mice. Under HFD conditions, body weight, body fat and liver fat content did not differ between genotypes. Yet, during intraperitoneal glucose tolerance tests (ipGTT) insulin excursions and HOMA-IR were lower in hKST-TG (4.42 ± 0.87 AU, WT vs. 2.20 ± 0.27 AU, hKST-TG, p < 0.05). Hyperinsulinemic euglycemic clamp studies with tracer-labeled glucose infusion confirmed improved insulin sensitivity by higher glucose infusion rates in hKST-TG mice (31.5 ± 1.78 mg/kg/min, hKST-TG vs. 18.1 ± 1.67 mg/kg/min, WT, p < 0.05). Improved insulin sensitivity was driven by reduced hepatic insulin resistance (clamp hepatic glucose output: 7.7 ± 1.9 mg/kg/min, hKST-TG vs 12.2 ± 0.8 mg/kg/min, WT, p < 0.05), providing evidence for direct insulin sensitizing effects of KST for the first time. Insulin sensitivity was differentially affected in skeletal muscle and adipose tissue. Mechanistically, we observed reduced Wnt signaling in the liver but not in skeletal muscle, which may explain the effect. CONCLUSIONS KST expression increases after weight loss in sWAT from people with obesity. Furthermore, human KST ameliorates diet-induced hepatic insulin resistance in mice, while differentially affecting skeletal muscle and adipose tissue insulin sensitivity. Thus, KST may be an interesting, yet challenging, therapeutic target for patients with obesity and insulin resistance.
Collapse
Affiliation(s)
- Leontine Sandforth
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tuebingen, Tuebingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Tuebingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sebastian Brachs
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Julia Reinke
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Section of Metabolic Vascular Medicine, Department of Medicine III, University Clinic Dresden, TU Dresden, Germany
| | - Diana Willmes
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Section of Metabolic Vascular Medicine, Department of Medicine III, University Clinic Dresden, TU Dresden, Germany
| | - Gencer Sancar
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tuebingen, Tuebingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Tuebingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Judith Seigner
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tuebingen, Tuebingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Tuebingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - David Juarez-Lopez
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tuebingen, Tuebingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Tuebingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Arvid Sandforth
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tuebingen, Tuebingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Tuebingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jeffrey D McBride
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Sven Haufe
- Department of Rehabilitation and Sports Medicine, Hannover Medical School (MHH), Hannover, Germany
| | - Jens Jordan
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany; Medical Faculty, University of Cologne, Cologne, Germany
| | - Andreas L Birkenfeld
- Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital of Tuebingen, Tuebingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Tuebingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Section of Metabolic Vascular Medicine, Department of Medicine III, University Clinic Dresden, TU Dresden, Germany; Department of Diabetes, Life Sciences & Medicine, Cardiovascular Medicine & Life Sciences, King's College London, UK.
| |
Collapse
|
3
|
Adedeji AO, Zhong F, Corpuz J, Hu F, Zhao X, Sangaraju D, Ruff CF, Dybdal N. Comparative Impact of Various Fasting Periods on the Welfare of Sprague-Dawley Rats With or Without Supplementation. Toxicol Pathol 2024; 52:21-34. [PMID: 38379371 DOI: 10.1177/01926233241230536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
In nonclinical toxicology studies, lab animals are fasted typically overnight, to reduce variability in some clinical pathology parameters. However, fasting adds undue stress, and this is particularly concerning in rodents given their fast metabolic rates. Furthermore, as rodents are nocturnal animals, an overnight fasting may cause a protracted negative metabolic state even when the fasting has technically ended, given their minimal activity and food consumption during the day. Therefore, to evaluate the impacts of different fasting durations (±DietGel supplementation) on rats' welfare, we assessed the traditional and ancillary clinical pathology parameters in Sprague-Dawley rats, along with body weight, organ weight, and histopathology. Although most endpoints were comparable between the different fasting durations (±DietGel supplementation), the long fasting times (≥8 hr) without DietGel supplementation caused significant decreases in body weight, liver weight, liver glycogen content, serum glucose, triglyceride, and creatinine concentrations-all findings suggestive of a negative energy balance that could impact animal welfare and consequently, data quality; while the short fasting time (4 hr) and DietGel supplementation were associated with higher triglycerides variability. Hence, we propose that short fasting time should be adequate for most toxicology studies in rats, and long fasting times should only be accommodated with scientific justification.
Collapse
Affiliation(s)
- Adeyemi O Adedeji
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| | - Fiona Zhong
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| | - Janice Corpuz
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| | - Fangyao Hu
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| | - Xiaofeng Zhao
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| | - Dewakar Sangaraju
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| | - Catherine F Ruff
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| | - Noel Dybdal
- Genentech, A Member of the Roche Group, South San Francisco, California, USA
| |
Collapse
|
4
|
Lyu K, Zhang D, Song J, Li X, Perry RJ, Samuel VT, Shulman GI. Short-term overnutrition induces white adipose tissue insulin resistance through sn-1,2-diacylglycerol/PKCε/insulin receptor Thr1160 phosphorylation. JCI Insight 2021; 6:139946. [PMID: 33411692 PMCID: PMC7934919 DOI: 10.1172/jci.insight.139946] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 12/29/2020] [Indexed: 12/17/2022] Open
Abstract
White adipose tissue (WAT) insulin action has critical anabolic function and is dysregulated in overnutrition. However, the mechanism of short-term high-fat diet-induced (HFD-induced) WAT insulin resistance (IR) is poorly understood. Based on recent evidences, we hypothesize that a short-term HFD causes WAT IR through plasma membrane (PM) sn-1,2-diacylglycerol (sn-1,2-DAG) accumulation, which promotes protein kinase C-ε (PKCε) activation to impair insulin signaling by phosphorylating insulin receptor (Insr) Thr1160. To test this hypothesis, we assessed WAT insulin action in 7-day HFD-fed versus regular chow diet-fed rats during a hyperinsulinemic-euglycemic clamp. HFD feeding caused WAT IR, reflected by impaired insulin-mediated WAT glucose uptake and lipolysis suppression. These changes were specifically associated with PM sn-1,2-DAG accumulation, higher PKCε activation, and impaired insulin-stimulated Insr Tyr1162 phosphorylation. In order to examine the role of Insr Thr1160 phosphorylation in mediating lipid-induced WAT IR, we examined these same parameters in InsrT1150A mice (mouse homolog for human Thr1160) and found that HFD feeding induced WAT IR in WT control mice but not in InsrT1150A mice. Taken together, these data demonstrate the importance of the PM sn-1,2-DAG/PKCε/Insr Thr1160 phosphorylation pathway in mediating lipid-induced WAT IR and represent a potential therapeutic target to improve WAT insulin sensitivity.
Collapse
Affiliation(s)
- Kun Lyu
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Dongyan Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Joongyu Song
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Xiruo Li
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Rachel J Perry
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Varman T Samuel
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA.,VA Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
5
|
Wang L, Sinnott-Armstrong N, Wagschal A, Wark AR, Camporez JP, Perry RJ, Ji F, Sohn Y, Oh J, Wu S, Chery J, Moud BN, Saadat A, Dankel SN, Mellgren G, Tallapragada DSP, Strobel SM, Lee MJ, Tewhey R, Sabeti PC, Schaefer A, Petri A, Kauppinen S, Chung RT, Soukas A, Avruch J, Fried SK, Hauner H, Sadreyev RI, Shulman GI, Claussnitzer M, Näär AM. A MicroRNA Linking Human Positive Selection and Metabolic Disorders. Cell 2020; 183:684-701.e14. [PMID: 33058756 PMCID: PMC8092355 DOI: 10.1016/j.cell.2020.09.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 05/08/2020] [Accepted: 09/03/2020] [Indexed: 01/09/2023]
Abstract
Positive selection in Europeans at the 2q21.3 locus harboring the lactase gene has been attributed to selection for the ability of adults to digest milk to survive famine in ancient times. However, the 2q21.3 locus is also associated with obesity and type 2 diabetes in humans, raising the possibility that additional genetic elements in the locus may have contributed to evolutionary adaptation to famine by promoting energy storage, but which now confer susceptibility to metabolic diseases. We show here that the miR-128-1 microRNA, located at the center of the positively selected locus, represents a crucial metabolic regulator in mammals. Antisense targeting and genetic ablation of miR-128-1 in mouse metabolic disease models result in increased energy expenditure and amelioration of high-fat-diet-induced obesity and markedly improved glucose tolerance. A thrifty phenotype connected to miR-128-1-dependent energy storage may link ancient adaptation to famine and modern metabolic maladaptation associated with nutritional overabundance.
Collapse
Affiliation(s)
- Lifeng Wang
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,These authors contributed equally,Present address: Cardiovascular & Metabolism, Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA 19477, USA
| | - Nasa Sinnott-Armstrong
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA,These authors contributed equally
| | - Alexandre Wagschal
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Present address: Vertex Pharmaceuticals, Watertown, MA 02472, USA
| | - Abigail R. Wark
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Joao-Paulo Camporez
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA,Present address: Ribeirao Preto School of Medicine, University of Sao Paulo, Sao Paulo 14049-90, Brazil
| | - Rachel J. Perry
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Fei Ji
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Yoojin Sohn
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Present address: Vanderbilt University, Nashville, TN 37235, USA
| | - Justin Oh
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Present address: Vertex Pharmaceuticals, Watertown, MA 02472, USA
| | - Su Wu
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Present address: Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Jessica Chery
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Present address: Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Bahareh Nemati Moud
- Else Kroener-Fresenius-Center of Nutritional Medicine, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Alham Saadat
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Simon N. Dankel
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, 5020 Bergen, Norway
| | - Gunnar Mellgren
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, 5020 Bergen, Norway
| | - Divya Sri Priyanka Tallapragada
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, 5020 Bergen, Norway
| | - Sophie Madlen Strobel
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, 80992 Munich, Germany
| | - Mi-Jeong Lee
- Obesity Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA,Present address: Department of Human Nutrition, Food and Animal Sciences, University of Hawaii, Honolulu, HI 96822, USA
| | - Ryan Tewhey
- Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA,Present address: The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Pardis C. Sabeti
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Anne Schaefer
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School ofMedicine atMount Sinai, New York, New York 10029, USA
| | - Andreas Petri
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, 2450 Copenhagen, Denmark
| | - Sakari Kauppinen
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, 2450 Copenhagen, Denmark
| | - Raymond T. Chung
- Liver Center, Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Alexander Soukas
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Department of Medicine, Center for Genomic Medicine and Diabetes Unit, Massachusetts General Hospital, Boston, MA 02114, USA,Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Joseph Avruch
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA,Department of Medicine, Harvard Medical School, Boston, MA 02114, USA,Diabetes unit, Medical Services, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Susan K. Fried
- Obesity Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA,Present address: Diabetes, Obesity and Metabolism Institute, Mt. Sinai School of Medicine, New York, NY 10029, USA
| | - Hans Hauner
- Else Kroener-Fresenius-Center of Nutritional Medicine, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany,Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, 80992 Munich, Germany
| | - Ruslan I. Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Gerald I. Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Melina Claussnitzer
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Anders M. Näär
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Present address: Department of Nutritional Sciences & Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA,Lead Contact,Correspondence: https://doi.org/10.1016/j.cell.2020.09.017
| |
Collapse
|
6
|
Wang Y, Nasiri AR, Damsky WE, Perry CJ, Zhang XM, Rabin-Court A, Pollak MN, Shulman GI, Perry RJ. Uncoupling Hepatic Oxidative Phosphorylation Reduces Tumor Growth in Two Murine Models of Colon Cancer. Cell Rep 2019; 24:47-55. [PMID: 29972790 DOI: 10.1016/j.celrep.2018.06.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 05/07/2018] [Accepted: 06/01/2018] [Indexed: 10/28/2022] Open
Abstract
Obesity is associated with colon cancer pathogenesis, but the underlying mechanism is actively debated. Here, we confirm that diet-induced obesity promotes tumor growth in two murine colon cancer models and show that this effect is reversed by an orally administered controlled-release mitochondrial protonophore (CRMP) that acts as a liver-specific uncoupler of oxidative phosphorylation. This agent lowered circulating insulin, and the reduction of tumor growth was abrogated by an insulin infusion raising plasma insulin to the level of high-fat-fed mice. We also demonstrate that hyperinsulinemia increases glucose uptake and oxidation in vivo in tumors and that CRMP reverses these effects. This study provides evidence that perturbations of whole-organism energy balance or hepatic energy metabolism can influence neoplastic growth. Furthermore, the data show that glucose uptake and utilization by cancers in vivo are not necessarily constitutively high but rather may vary according to the hormonal milieu.
Collapse
Affiliation(s)
- Yongliang Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ali R Nasiri
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - William E Damsky
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Dermatology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Curtis J Perry
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xian-Man Zhang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Aviva Rabin-Court
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Michael N Pollak
- Department of Oncology, McGill University, Montreal, Quebec H3T 1E2, Canada; Department of Medicine, McGill University, Montreal, Quebec H3T 1E2, Canada; Segal Cancer Centre, Jewish General Hospital, Montreal, Quebec H3T 1E2, Canada
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Rachel J Perry
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
7
|
Kovalenko EV, Mazina MY, Krasnov AN, Vorobyeva NE. The Drosophila nuclear receptors EcR and ERR jointly regulate the expression of genes involved in carbohydrate metabolism. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2019; 112:103184. [PMID: 31295549 DOI: 10.1016/j.ibmb.2019.103184] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/14/2019] [Accepted: 07/06/2019] [Indexed: 06/09/2023]
Abstract
The rate of carbohydrate metabolism is tightly coordinated with developmental transitions in Drosophila, and fluctuates depending on the requirements of a particular developmental stage. These successive metabolic switches result from changes in the expression levels of genes encoding glycolytic, tricarboxylic acid cycle (TCA), and oxidative phosphorylation enzymes. In this report, we describe a repressive action of ecdysone signaling on the expression of glycolytic genes and enzymes of glycogen metabolism in Drosophila development. The basis of this effect is an interaction between the ecdysone receptor (EcR) and the estrogen-related receptor (ERR), a specific regulator of the Drosophila glycolysis. We found an overlapping DNA-binding pattern for the EcR and ERR in the Drosophila S2 cells. EcR was detected at a subset of the ERR target genes responsible for carbohydrate metabolism. The 20-hydroxyecdysone treatment of both the Drosophila larvae and the S2 cells decreased transcriptional levels of ERR targets. We propose a joint action mode for both the EcR and ERR, for at least a subset of the glycolytic genes. We find that both receptors bind to the same regulatory regions and may form or be part of a joint transcriptional regulatory complex in the Drosophila S2 cells.
Collapse
Affiliation(s)
- Elena V Kovalenko
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Marina Yu Mazina
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Aleksey N Krasnov
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | | |
Collapse
|
8
|
Hollenbach M, Klöting N, Sommerer I, Lorenz J, Heindl M, Kern M, Mössner J, Blüher M, Hoffmeister A. p8 deficiency leads to elevated pancreatic beta cell mass but does not contribute to insulin resistance in mice fed with high-fat diet. PLoS One 2018; 13:e0201159. [PMID: 30040846 PMCID: PMC6057664 DOI: 10.1371/journal.pone.0201159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 07/10/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND p8 was initially described as being overexpressed in acute pancreatitis and encoding a ubiquitous stress protein. Analysis of insulin sensitivity and glucose tolerance in p8-knockout and haplodeficient mice revealed counterintuitive results. Thus, we determined glycemic control of p8 in mice fed with standard (SD) and high-fat diet (HFD). METHODS p8-/- and wild type (p8+/+) mice were used for analysis of glucagon (immunohistochemistry), insulin levels (ELISA) and beta cell mass. Hyperinsulinemic- euglycemic glucose clamp technique, i.p. glucose tolerance test (ipGTT), i.p. insulin tolerance test (ipITT) and metabolic chamber analysis were performed in SD (4% fat) and HFD (55% fat) groups. RESULTS p8-/- mice showed no differences in glucagon or insulin content but higher insulin secretion from pancreatic islets upon glucose stimulation. p8 deficiency resulted in elevated beta cell mass but was not associated with increased insulin resistance in ipGTT or ipITT. Glucose clamp tests also revealed no evidence of association of p8 deficiency with insulin resistance. Metabolic chamber analysis showed equal energy expenditure in p8-/- mice and wild type animals. CONCLUSION p8 depletion may contribute to glucose metabolism via stress-induced insulin production and elevated beta cell mass. Nevertheless, p8 knockout showed no impact on insulin resistance in SD and HFD-fed mice.
Collapse
Affiliation(s)
- Marcus Hollenbach
- Department of Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
- * E-mail:
| | - Nora Klöting
- IFB Adiposity Disease, University of Leipzig, Leipzig, Germany
| | - Ines Sommerer
- Department of Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Jana Lorenz
- Department of Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Mario Heindl
- Department of Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Matthias Kern
- German Diabetes Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Joachim Mössner
- Department of Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Department of Medicine, Neurology and Dermatology, Division of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Albrecht Hoffmeister
- Department of Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
9
|
Pathogenesis of hypothyroidism-induced NAFLD is driven by intra- and extrahepatic mechanisms. Proc Natl Acad Sci U S A 2017; 114:E9172-E9180. [PMID: 29073114 DOI: 10.1073/pnas.1707797114] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hypothyroidism, a metabolic disease characterized by low thyroid hormone (TH) and high thyroid-stimulating hormone (TSH) levels in the serum, is strongly associated with nonalcoholic fatty liver disease (NAFLD). Hypothyroidism-induced NAFLD has generally been attributed to reduced TH signaling in the liver with a consequent decrease in lipid utilization. Here, we found that mildly hypothyroid mice develop NAFLD without down-regulation of hepatic TH signaling or decreased hepatic lipid utilization. NAFLD was induced by impaired suppression of adipose tissue lipolysis due to decreased insulin secretion and to a reduced response of adipose tissue itself to insulin. This condition leads to increased shuttling of fatty acids (FAs) to the liver, where they are esterified and accumulated as triglycerides. Lipid accumulation in the liver induces hepatic insulin resistance, which leads to impaired suppression of endogenous glucose production after feeding. Hepatic insulin resistance, synergistically with lowered insulin secretion, increases serum glucose levels, which stimulates de novo lipogenesis (DNL) in the liver. Up-regulation of DNL also contributes to NAFLD. In contrast, severely hypothyroid mice show down-regulation of TH signaling in their livers and profound suppression of adipose tissue lipolysis, which decreases delivery of FAs to the liver. The resulting lack of substrates for triglyceride esterification protects severely hypothyroid mice against NAFLD. Our findings demonstrate that NAFLD occurs when TH levels are mildly reduced, but, paradoxically, not when they are severely reduced. Our results show that the pathogenesis of hypothyroidism-induced NAFLD is both intra- and extrahepatic; they also reveal key metabolic differences between mild and severe hypothyroidism.
Collapse
|
10
|
Yang J, Oh YT, Wan D, Watanabe RM, Hammock BD, Youn JH. Postprandial effect to decrease soluble epoxide hydrolase activity: roles of insulin and gut microbiota. J Nutr Biochem 2017; 49:8-14. [PMID: 28863368 DOI: 10.1016/j.jnutbio.2017.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/20/2017] [Accepted: 07/18/2017] [Indexed: 02/08/2023]
Abstract
Epoxides of free fatty acids (FFAs), especially epoxyeicosatrienoic acids (EETs), are lipid mediators with beneficial effects in metabolic and cardiovascular (CV) health. FFA epoxides are quickly metabolized to biologically less active diols by soluble epoxide hydrolase (sEH). Inhibition of sEH, which increases EET levels, improves glucose homeostasis and CV health and is proposed as an effective strategy for the treatment of diabetes and CV diseases. Here, we show evidence that sEH activity is profoundly reduced in postprandial states in rats; plasma levels of 17 sEH products (i.e., FFA diols), detected by targeted oxylipin analysis, all decreased after a meal. In addition, the ratios of sEH product to substrate (sEH P/S ratios), which may reflect sEH activity, decreased ~70% on average 2.5 h after a meal in rats (P<.01). To examine whether this effect was mediated by insulin action, a hyperinsulinemic-euglycemic clamp was performed for 2.5 h, and sEH P/S ratios were assessed before and after the clamp. The clamp resulted in small increases rather than decreases in sEH P/S ratios (P<.05), indicating that insulin cannot account for the postprandial decrease in sEH P/S ratios. Interestingly, in rats treated with antibiotics to deplete gut bacteria, the postprandial effect to decrease sEH P/S ratios was completely abolished, suggesting that a gut bacteria-derived factor(s) may be responsible for the effect. Further studies are warranted to identify such a factor(s) and elucidate the mechanism by which sEH activity (or sEH P/S ratio) is reduced in postprandial states.
Collapse
Affiliation(s)
- Jun Yang
- Department of Entomology and Nematology, University of California, Davis, CA, USA
| | - Young Taek Oh
- Department of Physiology and Biophysics, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Debin Wan
- Department of Entomology and Nematology, University of California, Davis, CA, USA
| | - Richard M Watanabe
- Department of Physiology and Biophysics, Keck School of Medicine of USC, Los Angeles, CA, USA; Department of Preventive Medicine, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology, University of California, Davis, CA, USA
| | - Jang H Youn
- Department of Physiology and Biophysics, Keck School of Medicine of USC, Los Angeles, CA, USA.
| |
Collapse
|
11
|
Oh YT, Oh HH, Nguyen AK, Choi CS, Youn JH. Circulating free fatty acids inhibit food intake in an oleate-specific manner in rats. Physiol Behav 2016; 167:194-201. [PMID: 27654062 DOI: 10.1016/j.physbeh.2016.09.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/31/2016] [Accepted: 09/14/2016] [Indexed: 10/21/2022]
Abstract
Previous rodent studies showed that when injected into the brain, free fatty acids (FFAs) reduced food intake in an oleate-specific manner. The present study was performed to test whether food intake is regulated by circulating FFAs in an oleate-specific manner. Male Wistar rats received an intravenous infusion of olive, safflower, or coconut oil (100mg/h), together with heparin, to raise circulating oleate, linoleate, or palmitate, respectively, and their effects on overnight food intake were evaluated. Compared to other oils, olive oil infusion showed a significantly greater effect to reduce food intake (P<0.01). Total caloric intake, the sum of the calories from the diet and infused oil, was significantly reduced with olive oil (P<0.01) but not with coconut or safflower oil infusion, suggesting an oleate-specific effect on caloric intake. To further test this idea, different groups of rats received an intravenous infusion of oleate, linoleate, or octanoate (0.5mg/h). Oleate infusion decreased overnight food intake by 26% (P<0.001), but no significant effect was seen with linoleate, octanoate, or vehicle infusion (P>0.05). The effects of olive oil or oleate infusion could not be explained by changes in plasma glucose, insulin, leptin, or total FFA levels. The olive oil effect on food intake was not reduced in vagotomized rats, suggesting that oleate sensing may not involve peripheral sensors. In contrast, olive oil's effect was attenuated in high-fat-fed rats, suggesting that this effect is regulated (or impaired) under physiological (or pathological) conditions. Taken together, the present study provides evidence that circulating oleate is sensed by the brain differentially from other FFAs to control feeding in rats.
Collapse
Affiliation(s)
- Young Taek Oh
- Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Hyun Hee Oh
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, Republic of Korea
| | - Anh-Khoi Nguyen
- Department of Exercise Sciences, University of Southern California, Los Angeles, CA, USA
| | - Cheol Soo Choi
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, Republic of Korea
| | - Jang H Youn
- Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
12
|
A potent and selective 11β-hydroxysteroid dehydrogenase type 1 inhibitor, SKI2852, ameliorates metabolic syndrome in diabetic mice models. Eur J Pharmacol 2015; 768:139-48. [PMID: 26519792 DOI: 10.1016/j.ejphar.2015.10.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 10/22/2015] [Accepted: 10/26/2015] [Indexed: 11/22/2022]
Abstract
11β-Hydroxysteroid dehydrogenase type 1 (11βHSD1) has been targeted for new drugs to treat type 2 diabetes and metabolic syndrome. In this study, we determined whether the inhibition of 11βHSD1 with a new selective inhibitor, SKI2852, could improve lipid profiles, glucose levels, and insulin sensitivity in type 2 diabetic and obese conditions. SKI2852 showed a potent inhibition of cortisone to cortisol conversion for over 80% in both liver and adipose tissue ex vivo from orally administered C57BL/6 mice, and in vivo analysis results were consistent with this. Repeated oral administrations of SKI2852 in diet-induced obesity (DIO) and ob/ob mice revealed a partially beneficial effect of SKI2852 in improving levels of cholesterols, triglycerides, free fatty acids, postprandial glucose, and/or blood hemoglobinA1c. SKI2852 significantly reduced body weight increase in ob/ob mice, and efficiently suppressed hepatic mRNA levels of gluconeogenic enzymes in DIO mice. Moreover, SKI2852 enhanced hepatic and whole body insulin sensitivities in hyperinsulinemic-euglycemic clamp experiment in DIO mice. In conclusion, these results indicate that selective and potent inhibition of 11βHSD1 by SKI2852, thus blockade of active glucocorticoid conversion, may improve many aspects of metabolic parameters in type 2 diabetes and metabolic diseases, mainly by inhibitions of hepatic gluconeogenesis and partial improvements of lipid profiles. Our study strongly support that SKI2852 may have a great potential as a novel candidate drug for the treatment of diabetes and metabolic diseases.
Collapse
|
13
|
Shin JH, Kim IY, Kim YN, Shin SM, Roh KJ, Lee SH, Sohn M, Cho SY, Lee SH, Ko CY, Kim HS, Choi CS, Bae YS, Seong JK. Obesity Resistance and Enhanced Insulin Sensitivity in Ahnak-/- Mice Fed a High Fat Diet Are Related to Impaired Adipogenesis and Increased Energy Expenditure. PLoS One 2015; 10:e0139720. [PMID: 26466345 PMCID: PMC4605776 DOI: 10.1371/journal.pone.0139720] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 09/16/2015] [Indexed: 11/18/2022] Open
Abstract
Objective Recent evidence has suggested that AHNAK expression is altered in obesity, although its role in adipose tissue development remains unclear. The objective of this study was to determine the molecular mechanism by which Ahnak influences adipogenesis and glucose homeostasis. Design We investigated the in vitro role of AHNAK in adipogenesis using adipose-derived mesenchymal stem cells (ADSCs) and C3H10T1/2 cells. AHNAK-KO male mice were fed a high-fat diet (HFD; 60% calories from fat) and examined for glucose and insulin tolerances, for body fat compositions, and by hyperinsulinemic-euglycemic clamping. Energy expenditures were assessed using metabolic cages and by measuring the expression levels of genes involved in thermogenesis in white or brown adipose tissues. Results Adipogenesis in ADSCs was impaired in AHNAK-KO mice. The loss of AHNAK led to decreased BMP4/SMAD1 signaling, resulting in the downregulation of key regulators of adipocyte differentiation (P<0.05). AHNAK directly interacted with SMAD1 on the Pparγ2 promoter. Concomitantly, HFD-fed AHNAK-KO mice displayed reduced hepatosteatosis and improved metabolic profiles, including improved glucose tolerance (P<0.001), enhanced insulin sensitivity (P<0.001), and increased energy expenditure (P<0.05), without undergoing alterations in food intake and physical activity. Conclusion AHNAK plays a crucial role in body fat accumulation by regulating adipose tissue development via interaction with the SMAD1 protein and can be involved in metabolic homeostasis.
Collapse
Affiliation(s)
- Jae Hoon Shin
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, and BK21 Program for Veterinary Science, Seoul National University, Seoul, South Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea
| | - Il Yong Kim
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, and BK21 Program for Veterinary Science, Seoul National University, Seoul, South Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea
| | - Yo Na Kim
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, and BK21 Program for Veterinary Science, Seoul National University, Seoul, South Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea
| | - Sun Mee Shin
- Division of Life Sciences, Ewha Womans University, Seoul, South Korea
| | - Kyung Jin Roh
- Lee Gil Ya Cancer and Diabetes Institute and Division of Endocrinology Gil Medical Center, Gachon University of Medicine and Science, Incheon, South Korea
| | - Seo Hyun Lee
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, and BK21 Program for Veterinary Science, Seoul National University, Seoul, South Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea
| | - Mira Sohn
- Division of Life Sciences, Ewha Womans University, Seoul, South Korea
| | - Soo Young Cho
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, and BK21 Program for Veterinary Science, Seoul National University, Seoul, South Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea
| | - Sang Hyuk Lee
- Ewha Research Center for Systems Biology, Ewha Womans University, Seoul, South Korea
| | - Chang-Yong Ko
- Department of Biomedical Engineering, College of Health Science, Institute of Medical Engineering, Yonsei University, Wonju, South Korea
| | - Han-Sung Kim
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, and BK21 Program for Veterinary Science, Seoul National University, Seoul, South Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea
| | - Cheol Soo Choi
- Lee Gil Ya Cancer and Diabetes Institute and Division of Endocrinology Gil Medical Center, Gachon University of Medicine and Science, Incheon, South Korea
| | - Yun Soo Bae
- Division of Life Sciences, Ewha Womans University, Seoul, South Korea
- * E-mail: (JKS); (YSB)
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, and BK21 Program for Veterinary Science, Seoul National University, Seoul, South Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea
- Interdisciplinary Program for Bioinformatics, Program for Cancer Biology and BIO-MAX Institute, Seoul National University, Seoul, South Korea
- * E-mail: (JKS); (YSB)
| |
Collapse
|
14
|
Short-term food restriction followed by controlled refeeding promotes gorging behavior, enhances fat deposition, and diminishes insulin sensitivity in mice. J Nutr Biochem 2015; 26:721-8. [PMID: 25913018 DOI: 10.1016/j.jnutbio.2015.01.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/08/2015] [Accepted: 01/23/2015] [Indexed: 01/08/2023]
Abstract
Rodents are commonly used in food restriction refeeding studies to investigate weight regain. Mice that are rationed food every 24 h may consume all allocated food in a short time (gorge) and therefore undergo a brief well-fed period followed by an extended fasted period until the next day's food allotment. These exaggerated metabolic states are not typical in mice fed ad libitum (nibbling). The aim of the current study was to elucidate the intraday and cumulative metabolic consequences of gorging (induced by food restriction) in mice during controlled refeeding. Accordingly, following a temporary food restriction, mice were fed rations similar to intakes of controls fed ad libitum. Temporary food restriction initiated gorging behavior that persisted during refeeding; consequently, metabolism-related measurements were obtained in the gorging mice during their daily fed and fasted metabolic states. Robust differences in adipose tissue lipogenic and inflammatory gene expression were found in the gorging mice by metabolic state (fed versus fasted). Additionally, despite a reduced cumulative food intake compared to mice fed ad libitum, restriction-induced gorging mice had increased intraabdominal fat accumulation, diminished hepatic and peripheral insulin sensitivity, and a gene expression profile favoring lipid deposition. Our findings highlight the intraday differences in gene expression in gorging mice before and after feeding that confound comparisons with mice fed ad libitum, or nibbling. The present study also provides evidence that weight regain following food restriction is associated with cumulative metabolic and behavioral abnormalities in mice.
Collapse
|
15
|
Li R, Xu X, Chen C, Wang Y, Gruzdev A, Zeldin DC, Wang DW. CYP2J2 attenuates metabolic dysfunction in diabetic mice by reducing hepatic inflammation via the PPARγ. Am J Physiol Endocrinol Metab 2015; 308:E270-82. [PMID: 25389363 PMCID: PMC4329496 DOI: 10.1152/ajpendo.00118.2014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Epoxyeicosatrienoic acids (EETs) and arachidonic acid-derived cytochrome P450 (CYP) epoxygenase metabolites have diverse biological effects, including anti-inflammatory properties in the vasculature. Increasing evidence suggests that inflammation in type 2 diabetes is a key component in the development of insulin resistance. In this study, we investigated whether CYP epoxygenase expression and exogenous EETs can attenuate insulin resistance in diabetic db/db mice and in cultured hepatic cells (HepG2). In vivo, CYP2J2 expression and the accompanying increase in EETs attenuated insulin resistance, as determined by plasma glucose levels, glucose tolerance test, insulin tolerance test, and hyperinsulinemic euglycemic clamp studies. CYP2J2 expression reduced the production of proinflammatory cytokines in liver, including CRP, IL-6, IL-1β, and TNFα, and decreased the infiltration of macrophages in liver. CYP2J2 expression also decreased activation of proinflammatory signaling cascades by decreasing NF-κB and MAPK activation in hepatocytes. Interestingly, CYP2J2 expression and exogenous EET treatment increased glucose uptake and activated the insulin-signaling cascade both in vivo and in vitro, suggesting that CYP2J2 metabolites play a role in glucose homeostasis. Furthermore, CYP2J2 expression upregulated PPARγ, which has been shown to induce adipogenesis, which attenuates dyslipidemias observed in diabetes. All of the findings suggest that CYP2J2 expression attenuates the diabetic phenotype and insulin resistance via inhibition of NF-κB and MAPK signaling pathways and activation of PPARγ.
Collapse
Affiliation(s)
- Rui Li
- Departments of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Xizhen Xu
- Departments of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Chen Chen
- Departments of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Yan Wang
- Departments of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Artiom Gruzdev
- Division of Intramural Research, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina
| | - Dao Wen Wang
- Departments of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| |
Collapse
|
16
|
Kern M, Kosacka J, Hesselbarth N, Brückner J, Heiker JT, Flehmig G, Klöting I, Kovacs P, Matz-Soja M, Gebhardt R, Krohn K, Sales S, Abshagen K, Shevchenko A, Stumvoll M, Blüher M, Klöting N. Liver-restricted Repin1 deficiency improves whole-body insulin sensitivity, alters lipid metabolism, and causes secondary changes in adipose tissue in mice. Diabetes 2014; 63:3295-309. [PMID: 24760138 DOI: 10.2337/db13-0933] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Replication initiator 1 (Repin1) is a zinc finger protein highly expressed in liver and adipose tissue and maps within a quantitative trait locus (QTL) for body weight and triglyceride (TG) levels in the rat. The QTL has further been supported as a susceptibility locus for dyslipidemia and related metabolic disorders in congenic and subcongenic rat strains. Here, we elucidated the role of Repin1 in lipid metabolism in vivo. We generated a liver-specific Repin1 knockout mouse (LRep1(-/-)) and systematically characterized the consequences of Repin1 deficiency in the liver on body weight, glucose and lipid metabolism, liver lipid patterns, and protein/mRNA expression. Hyperinsulinemic-euglycemic clamp studies revealed significantly improved whole-body insulin sensitivity in LRep1(-/-) mice, which may be due to significantly lower TG content in the liver. Repin1 deficiency causes significant changes in potential downstream target molecules including Cd36, Pparγ, Glut2 protein, Akt phosphorylation, and lipocalin2, Vamp4, and Snap23 mRNA expression. Mice with hepatic deletion of Repin1 display secondary changes in adipose tissue function, which may be mediated by altered hepatic expression of lipocalin2 or chemerin. Our findings indicate that Repin1 plays a role in insulin sensitivity and lipid metabolism by regulating key genes of glucose and lipid metabolism.
Collapse
Affiliation(s)
- Matthias Kern
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Joanna Kosacka
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | | | - Julia Brückner
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - John T Heiker
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Gesine Flehmig
- IFB AdiposityDiseases, University of Leipzig, Leipzig, Germany
| | - Ingrid Klöting
- Department of Laboratory Animal Science, University of Greifswald, Karlsburg, Germany
| | - Peter Kovacs
- IFB AdiposityDiseases, University of Leipzig, Leipzig, Germany
| | - Madlen Matz-Soja
- Institute of Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Rolf Gebhardt
- Institute of Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Knut Krohn
- Interdisciplinary Center for Clinical Research, Core Unit DNA Technologies, University of Leipzig, Leipzig, Germany
| | - Susanne Sales
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Kerstin Abshagen
- Institute for Experimental Surgery, Rostock University Medical School, Rostock, Germany
| | - Andrej Shevchenko
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Michael Stumvoll
- Department of Medicine, University of Leipzig, Leipzig, Germany IFB AdiposityDiseases, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany IFB AdiposityDiseases, University of Leipzig, Leipzig, Germany
| | - Nora Klöting
- Department of Medicine, University of Leipzig, Leipzig, Germany IFB AdiposityDiseases, University of Leipzig, Leipzig, Germany
| |
Collapse
|
17
|
In-depth metabolic phenotyping of genetically engineered mouse models in obesity and diabetes. Mamm Genome 2014; 25:508-21. [PMID: 24792749 DOI: 10.1007/s00335-014-9520-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 04/10/2014] [Indexed: 01/09/2023]
Abstract
The world-wide prevalence of obesity and diabetes has increased sharply during the last two decades. Accordingly, the metabolic phenotyping of genetically engineered mouse models is critical for evaluating the functional roles of target genes in obesity and diabetes, and for developing new therapeutic targets. In this review, we discuss the practical meaning of metabolic phenotyping, the strategy of choosing appropriate tests, and considerations when designing and performing metabolic phenotyping in mice.
Collapse
|
18
|
Oh YT, Kim J, Kang I, Youn JH. Regulation of hypothalamic-pituitary-adrenal axis by circulating free fatty acids in male Wistar rats: role of individual free fatty acids. Endocrinology 2014; 155:923-31. [PMID: 24424035 PMCID: PMC3929730 DOI: 10.1210/en.2013-1700] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We previously showed that a fall in the plasma free fatty acid (FFA) level increases plasma corticosterone levels in rats by activating the hypothalamic-pituitary-adrenal axis. In the present study, we tested whether this regulation is mediated by specific or all species of FFAs. Nicotinic acid (NA) (30 μmol/h) was infused in rats to decrease plasma FFAs and increase plasma ACTH and corticosterone. The NA infusion was combined with an infusion of lipids with different FFA compositions to selectively prevent falls in individual FFA levels; coconut, olive, and safflower oils (n = 7 for each), which are predominantly (>70%) composed of saturated, monounsaturated (oleic acid), and polyunsaturated (linoleic acid) FFAs, respectively, were used. At an infusion rate (0.1 g/h) that only partially prevented a fall in the total FFA level, coconut oil, but not olive or safflower oil, completely prevented NA-induced increases in plasma ACTH or corticosterone, suggesting that these responses are mainly mediated by saturated FFAs. In addition, quantification of individual FFA species in the blood using FFA-specific fluorescent probes revealed that plasma corticosterone and ACTH correlated significantly with plasma palmitate but not with other FFAs, such as oleate, linoleate, or arachidonate. Taken together, our data suggest that the regulation of the hypothalamic-pituitary-adrenal axis by FFAs is mainly mediated by the saturated fatty acid palmitate, but not by unsaturated fatty acids, such as oleate and linoleate.
Collapse
Affiliation(s)
- Young Taek Oh
- Department of Physiology and Biophysics (Y.T.O., J.K., J.H.Y.), University of Southern California, Keck School of Medicine, Los Angeles, California 90089; and Department of Biochemistry and Molecular Biology (I.K., J.H.Y.), Kyung Hee University, School of Medicine, Seoul 130-701, Korea
| | | | | | | |
Collapse
|
19
|
Camporez JPG, Jornayvaz FR, Petersen MC, Pesta D, Guigni BA, Serr J, Zhang D, Kahn M, Samuel VT, Jurczak MJ, Shulman GI. Cellular mechanisms by which FGF21 improves insulin sensitivity in male mice. Endocrinology 2013; 154:3099-109. [PMID: 23766126 PMCID: PMC3749479 DOI: 10.1210/en.2013-1191] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fibroblast growth factor 21 (FGF21) is a potent regulator of glucose and lipid metabolism and is currently being pursued as a therapeutic agent for insulin resistance and type 2 diabetes. However, the cellular mechanisms by which FGF21 modifies insulin action in vivo are unclear. To address this question, we assessed insulin action in regular chow- and high-fat diet (HFD)-fed wild-type mice chronically infused with FGF21 or vehicle. Here, we show that FGF21 administration results in improvements in both hepatic and peripheral insulin sensitivity in both regular chow- and HFD-fed mice. This improvement in insulin responsiveness in FGF21-treated HFD-fed mice was associated with decreased hepatocellular and myocellular diacylglycerol content and reduced protein kinase Cε activation in liver and protein kinase Cθ in skeletal muscle. In contrast, there were no effects of FGF21 on liver or muscle ceramide content. These effects may be attributed, in part, to increased energy expenditure in the liver and white adipose tissue. Taken together, these data provide a mechanism by which FGF21 protects mice from lipid-induced liver and muscle insulin resistance and support its development as a novel therapy for the treatment of nonalcoholic fatty liver disease, insulin resistance, and type 2 diabetes.
Collapse
MESH Headings
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/surgery
- Animals
- Cells, Cultured
- Diet, High-Fat/adverse effects
- Drug Implants
- Energy Metabolism/drug effects
- Fibroblast Growth Factors/administration & dosage
- Fibroblast Growth Factors/metabolism
- Fibroblast Growth Factors/therapeutic use
- Glucose Intolerance/drug therapy
- Glucose Intolerance/etiology
- Glucose Intolerance/metabolism
- Glucose Intolerance/pathology
- Humans
- Infusions, Subcutaneous
- Insulin Resistance
- Isoenzymes/metabolism
- Lipectomy
- Lipid Metabolism/drug effects
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Protein Kinase C/metabolism
- Protein Kinase C-epsilon/metabolism
- Protein Kinase C-theta
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/metabolism
- Recombinant Proteins/therapeutic use
Collapse
Affiliation(s)
- João Paulo G Camporez
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06536-9812, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Ma B, Xiong X, Chen C, Li H, Xu X, Li X, Li R, Chen G, Dackor RT, Zeldin DC, Wang DW. Cardiac-specific overexpression of CYP2J2 attenuates diabetic cardiomyopathy in male streptozotocin-induced diabetic mice. Endocrinology 2013; 154:2843-56. [PMID: 23696562 PMCID: PMC3713213 DOI: 10.1210/en.2012-2166] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cytochrome P450 (CYP) epoxygenases metabolize arachidonic acid to biologically active cis-epoxyeicosatrienoic acids, which have potent vasodilatory, antiinflammatory, antiapoptotic, and antidiabetes properties. Here, we showed the effects of cardiac-specific overexpression of CYP epoxygenase 2J2 (CYP2J2) on diabetic cardiomyopathy and insulin resistance in high-fat (HF) diet fed, low-dose streptozotocin-treated mice. Diabetic cardiomyopathy was induced by HF and streptozotocin in cardiac-specific CYP2J2 transgenic mice. Physiological parameters and systemic metabolic parameters were monitored using ELISA kits. Intraperitoneal injection glucose tolerance test and hyperinsulinemic-euglycemic clamp study were implied to indicate insulin resistance. Cardiac function was assessed by echocardiography and Millar catheter system. Real-time PCR and Western blotting were used in signal pathway detection. αMHC-CYP2J2 transgenic mice showed significantly lower plasma glucose and insulin levels, improved glucose tolerance, and increased cardiac glucose uptake. Furthermore, αMHC-CYP2J2 transgenic mice were significantly protected from HF-streptozotocin-induced diabetic cardiomyopathy. Strikingly, CYP2J2 overexpression attenuated myocardial hypertrophy induced by diabetes. We conclude that cardiac-specific overexpression of CYP2J2 significantly protects against diabetic cardiomyopathy, which may be due to improved cardiac insulin resistance, glucose uptake, and reversal of cardiac hypertrophy. Relevant mechanisms may include up-regulation of peroxisome proliferator-activated receptor γ, activation of insulin receptor and AMP-activated protein kinase signaling pathways, and inhibition of nuclear factor of activated T cells c3 signal by enhanced atrial natriuretic peptide production. These results suggest that CYP2J2 epoxygenase metabolites likely play an important role in plasma glucose homeostasis, and enhancement of epoxyeicosatrienoic acids activation may serve as an effective therapeutic strategy to prevent diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Ben Ma
- The Institute of Hypertension and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People’s Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Löffler MG, Birkenfeld AL, Philbrick KM, Belman JP, Habtemichael EN, Booth CJ, Castorena CM, Choi CS, Jornayvaz FR, Gassaway BM, Lee HY, Cartee GD, Philbrick W, Shulman GI, Samuel VT, Bogan JS. Enhanced fasting glucose turnover in mice with disrupted action of TUG protein in skeletal muscle. J Biol Chem 2013; 288:20135-50. [PMID: 23744065 DOI: 10.1074/jbc.m113.458075] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Insulin stimulates glucose uptake in 3T3-L1 adipocytes in part by causing endoproteolytic cleavage of TUG (tether containing a ubiquitin regulatory X (UBX) domain for glucose transporter 4 (GLUT4)). Cleavage liberates intracellularly sequestered GLUT4 glucose transporters for translocation to the cell surface. To test the role of this regulation in muscle, we used mice with muscle-specific transgenic expression of a truncated TUG fragment, UBX-Cter. This fragment causes GLUT4 translocation in unstimulated 3T3-L1 adipocytes. We predicted that transgenic mice would have GLUT4 translocation in muscle during fasting. UBX-Cter expression caused depletion of PIST (PDZ domain protein interacting specifically with TC10), which transmits an insulin signal to TUG. Whereas insulin stimulated TUG proteolysis in control muscles, proteolysis was constitutive in transgenic muscles. Fasting transgenic mice had decreased plasma glucose and insulin concentrations compared with controls. Whole-body glucose turnover was increased during fasting but not during hyperinsulinemic clamp studies. In muscles with the greatest UBX-Cter expression, 2-deoxyglucose uptake during fasting was similar to that in control muscles during hyperinsulinemic clamp studies. Fasting transgenic mice had increased muscle glycogen, and GLUT4 targeting to T-tubule fractions was increased 5.7-fold. Whole-body oxygen consumption (VO2), carbon dioxide production (VCO2), and energy expenditure were increased by 12-13%. After 3 weeks on a high fat diet, the decreased fasting plasma glucose in transgenic mice compared with controls was more marked, and increased glucose turnover was not observed; the transgenic mice continued to have an increased metabolic rate. We conclude that insulin stimulates TUG proteolysis to translocate GLUT4 in muscle, that this pathway impacts systemic glucose homeostasis and energy metabolism, and that the effects of activating this pathway are maintained during high fat diet-induced insulin resistance in mice.
Collapse
Affiliation(s)
- Michael G Löffler
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8020, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Burghoff S, Flögel U, Bongardt S, Burkart V, Sell H, Tucci S, Ikels K, Eberhard D, Kern M, Klöting N, Eckel J, Schrader J. Deletion of CD73 promotes dyslipidemia and intramyocellular lipid accumulation in muscle of mice. Arch Physiol Biochem 2013; 119:39-51. [PMID: 23398498 DOI: 10.3109/13813455.2012.755547] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CONTEXT CD73 converts extracellular AMP to adenosine which is well known to inhibit lipolysis. It is unknown, however, whether adenosine formed directly by CD73 is functionally relevant in this process. OBJECTIVE We therefore explored the effect of CD73-derived adenosine on body fat of aged mice. RESULTS In lean mice, extracellular adenosine formation by adipocytes is dependent on CD73. High fat diet down-regulates the expression of CD73 in wildtype mice similar to ob/ob mice. Transgenic mice chronically lacking CD73 (CD73(-/-)) gain significantly less body weight and show decreased superficial white fat content as well as increased serum free fatty acids and triglycerides. In addition, intramyocellular lipid levels are significantly increased. This phenotype is accompanied by an increase in blood glucose and serum insulin levels although insulin secretion and the level of insulin degrading enzyme are unaltered. Additionally, insulin-induced Akt phosphorylation is reduced in skeletal muscle of CD73(-/-) mice. CONCLUSION CD73-derived adenosine is functionally involved in body fat homeostasis.
Collapse
Affiliation(s)
- Sandra Burghoff
- Institute of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Oh YT, Oh KS, Kang I, Youn JH. A Fall in plasma free fatty acid (FFA) level activates the hypothalamic-pituitary-adrenal axis independent of plasma glucose: evidence for brain sensing of circulating FFA. Endocrinology 2012; 153:3587-92. [PMID: 22669895 PMCID: PMC3404348 DOI: 10.1210/en.2012-1330] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The brain responds to a fall in blood glucose by activating neuroendocrine mechanisms for its restoration. It is unclear whether the brain also responds to a fall in plasma free fatty acids (FFA) to activate mechanisms for its restoration. We examined whether lowering plasma FFA increases plasma corticosterone or catecholamine levels and, if so, whether the brain is involved in these responses. Plasma FFA levels were lowered in rats with three independent antilipolytic agents: nicotinic acid (NA), insulin, and the A1 adenosine receptor agonist SDZ WAG 994 with plasma glucose clamped at basal levels. Lowering plasma FFA with these agents all increased plasma corticosterone, but not catecholamine, within 1 h, accompanied by increases in plasma ACTH. These increases in ACTH or corticosterone were abolished when falls in plasma FFA were prevented by Intralipid during NA or insulin infusion. In addition, the NA-induced increases in plasma ACTH were completely prevented by administration of SSR149415, an arginine vasopressin receptor antagonist, demonstrating that the hypothalamus is involved in these responses. Taken together, the present data suggest that the brain may sense a fall in plasma FFA levels and activate the hypothalamic-pituitary-adrenal axis to increase plasma ACTH and corticosterone, which would help restore FFA levels. Thus, the brain may be involved in the sensing and control of circulating FFA levels.
Collapse
Affiliation(s)
- Young Taek Oh
- Department of Physiology and Biophysics, University of Southern California, Keck School of Medicine, 1333 San Pablo Street, Mudd Memorial Research Building 626, Los Angeles, California 90089-9142, USA
| | | | | | | |
Collapse
|
24
|
Jurczak MJ, Lee AH, Jornayvaz FR, Lee HY, Birkenfeld AL, Guigni BA, Kahn M, Samuel VT, Glimcher LH, Shulman GI. Dissociation of inositol-requiring enzyme (IRE1α)-mediated c-Jun N-terminal kinase activation from hepatic insulin resistance in conditional X-box-binding protein-1 (XBP1) knock-out mice. J Biol Chem 2011; 287:2558-67. [PMID: 22128176 PMCID: PMC3268415 DOI: 10.1074/jbc.m111.316760] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatic insulin resistance has been attributed to both increased endoplasmic reticulum (ER) stress and accumulation of intracellular lipids, specifically diacylglycerol (DAG). The ER stress response protein, X-box-binding protein-1 (XBP1), was recently shown to regulate hepatic lipogenesis, suggesting that hepatic insulin resistance in models of ER stress may result from defective lipid storage, as opposed to ER-specific stress signals. Studies were designed to dissociate liver lipid accumulation and activation of ER stress signaling pathways, which would allow us to delineate the individual contributions of ER stress and hepatic lipid content to the pathogenesis of hepatic insulin resistance. Conditional XBP1 knock-out (XBP1Δ) and control mice were fed fructose chow for 1 week. Determinants of whole-body energy balance, weight, and composition were determined. Hepatic lipids including triglyceride, DAGs, and ceramide were measured, alongside markers of ER stress. Whole-body and tissue-specific insulin sensitivity were determined by hyperinsulinemic-euglycemic clamp studies. Hepatic ER stress signaling was increased in fructose chow-fed XBP1Δ mice as reflected by increased phosphorylated eIF2α, HSPA5 mRNA, and a 2-fold increase in hepatic JNK activity. Despite JNK activation, XBP1Δ displayed increased hepatic insulin sensitivity during hyperinsulinemic-euglycemic clamp studies, which was associated with increased insulin-stimulated IRS2 tyrosine phosphorylation, reduced hepatic DAG content, and reduced PKCε activity. These studies demonstrate that ER stress and IRE1α-mediated JNK activation can be disassociated from hepatic insulin resistance and support the hypothesis that hepatic insulin resistance in models of ER stress may be secondary to ER stress modulation of hepatic lipogenesis.
Collapse
Affiliation(s)
- Michael J Jurczak
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06536-8012, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ning J, Hong T, Yang X, Mei S, Liu Z, Liu HY, Cao W. Insulin and insulin signaling play a critical role in fat induction of insulin resistance in mouse. Am J Physiol Endocrinol Metab 2011; 301:E391-401. [PMID: 21586696 PMCID: PMC3154527 DOI: 10.1152/ajpendo.00164.2011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The primary player that induces insulin resistance has not been established. Here, we studied whether or not fat can cause insulin resistance in the presence of insulin deficiency. Our results showed that high-fat diet (HFD) induced insulin resistance in C57BL/6 (B6) mice. The HFD-induced insulin resistance was prevented largely by the streptozotocin (STZ)-induced moderate insulin deficiency. The STZ-induced insulin deficiency prevented the HFD-induced ectopic fat accumulation and oxidative stress in liver and gastrocnemius. The STZ-induced insulin deficiency prevented the HFD- or insulin-induced increase in hepatic expression of long-chain acyl-CoA synthetases (ACSL), which are necessary for fatty acid activation. HFD increased mitochondrial contents of long-chain acyl-CoAs, whereas it decreased mitochondrial ADP/ATP ratio, and these HFD-induced changes were prevented by the STZ-induced insulin deficiency. In cultured hepatocytes, we observed that expressions of ACSL1 and -5 were stimulated by insulin signaling. Results in cultured cells also showed that blunting insulin signaling by the PI3K inhibitor LY-294002 prevented fat accumulation, oxidative stress, and insulin resistance induced by the prolonged exposure to either insulin or oleate plus sera that normally contain insulin. Finally, knockdown of the insulin receptor prevented the oxidative stress and insulin resistance induced by the prolonged exposure to insulin or oleate plus sera. Together, our results show that insulin and insulin signaling are required for fat induction of insulin resistance in mice and cultured mouse hepatocytes.
Collapse
Affiliation(s)
- Jie Ning
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill,Chapel Hill, NC 27559, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Purcell SH, Aerni-Flessner LB, Willcockson AR, Diggs-Andrews KA, Fisher SJ, Moley KH. Improved insulin sensitivity by GLUT12 overexpression in mice. Diabetes 2011; 60:1478-82. [PMID: 21441439 PMCID: PMC3292321 DOI: 10.2337/db11-0033] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Evidence suggests that insulin-sensitive glucose transporters (GLUTs) other than GLUT4 may exist. To investigate whether GLUT12 may represent another insulin-sensitive GLUT, transgenic (TG) mice that overexpress GLUT12 were characterized. RESEARCH DESIGN AND METHODS TG mice that overexpressed GLUT12 under a β-actin promoter were generated. Glucose metabolism in TG and wild-type control mice was compared using glucose and insulin tolerance tests and hyperinsulinemic-euglycemic clamps. In addition, basal and insulin-stimulated glucose clearance rates into insulin-sensitive peripheral tissues were measured using [(3)H]-2-deoxy-D-glucose. RESULTS GLUT12 was overexpressed by 40-75% in TG compared with wild-type mice in insulin-sensitive tissues with no change in GLUT4 content. Body weight and fasting blood glucose did not differ between wild-type and TG mice; however, insulin concentrations were reduced in TG mice. Enhanced oral glucose tolerance was noted in TG mice by a reduced blood glucose excursion compared with wild-type mice (P < 0.05). Enhanced insulin sensitivity was noted by a greater decrease in blood glucose in TG mice during insulin tolerance testing. Hyperinsulinemic-euglycemic clamps confirmed enhanced insulin sensitivity in GLUT12-overexpressing mice (P < 0.01). Tissues of TG mice exhibited normal basal glucose clearance rates; however, under insulin-stimulated conditions, glucose clearance was significantly increased (P < 0.01) in tissues of TG mice. CONCLUSIONS Increased expression of GLUT12 results in improved whole-body insulin sensitivity mediated by an increased glucose clearance rate in insulin-responsive tissues under insulin-stimulated, but not basal, conditions. These findings provide evidence that GLUT12 represents a novel, second insulin-sensitive GLUT.
Collapse
Affiliation(s)
- Scott H. Purcell
- Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, Missouri
| | - Lauren B. Aerni-Flessner
- Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, Missouri
| | - Alexandra R. Willcockson
- Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, Missouri
| | - Kelly A. Diggs-Andrews
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Simon J. Fisher
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Kelle H. Moley
- Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, Missouri
- Corresponding author: Kelle H. Moley,
| |
Collapse
|
27
|
Liu L, Shi X, Choi CS, Shulman GI, Klaus K, Nair KS, Schwartz GJ, Zhang Y, Goldberg IJ, Yu YH. Paradoxical coupling of triglyceride synthesis and fatty acid oxidation in skeletal muscle overexpressing DGAT1. Diabetes 2009; 58:2516-24. [PMID: 19675136 PMCID: PMC2768165 DOI: 10.2337/db08-1096] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Transgenic expression of diacylglycerol acyltransferase-1 (DGAT1) in skeletal muscle leads to protection against fat-induced insulin resistance despite accumulation of intramuscular triglyceride, a phenomenon similar to what is known as the "athlete paradox." The primary objective of this study is to determine how DGAT1 affects muscle fatty acid oxidation in relation to whole-body energy metabolism and insulin sensitivity. RESEARCH DESIGN AND METHODS We first quantified insulin sensitivity and the relative tissue contributions to the improved whole-body insulin sensitivity in muscle creatine kisase (MCK)-DGAT1 transgenic mice by hyperinsulinemic-euglycemic clamps. Metabolic consequences of DGAT1 overexpression in skeletal muscles were determined by quantifying triglyceride synthesis/storage (anabolic) and fatty acid oxidation (catabolic), in conjunction with gene expression levels of representative marker genes in fatty acid metabolism. Whole-body energy metabolism including food consumption, body weights, oxygen consumption, locomotor activity, and respiration exchange ratios were determined at steady states. RESULTS MCK-DGAT1 mice were protected against muscle lipoptoxicity, although they remain susceptible to hepatic lipotoxicity. While augmenting triglyceride synthesis, DGAT1 overexpression also led to increased muscle mitochondrial fatty acid oxidation efficiency, as compared with wild-type muscles. On a high-fat diet, MCK-DGAT1 mice displayed higher basal metabolic rates and 5-10% lower body weights compared with wild-type littermates, whereas food consumption was not different. CONCLUSIONS DGAT1 overexpression in skeletal muscle led to parallel increases in triglyceride synthesis and fatty acid oxidation. Seemingly paradoxical, this phenomenon is characteristic of insulin-sensitive myofibers and suggests that DGAT1 plays an active role in metabolic "remodeling" of skeletal muscle coupled with insulin sensitization.
Collapse
Affiliation(s)
- Li Liu
- Department of Medicine, Preventive Medicine and Nutrition, Columbia University, New York, New York
| | - Xiaojing Shi
- Department of Medicine, Preventive Medicine and Nutrition, Columbia University, New York, New York
| | - Cheol Soo Choi
- Departments of Internal Medicine and Cellular and Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut
| | - Gerald I. Shulman
- Departments of Internal Medicine and Cellular and Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut
| | - Katherine Klaus
- Endocrine Research Unit and Department of Laboratory Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - K. Sreekumaran Nair
- Endocrine Research Unit and Department of Laboratory Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Gary J. Schwartz
- Department of Medicine & Neuroscience, Diabetes Research and Training Center, Albert Einstein College of Medicine, New York, New York
| | - Yiying Zhang
- Department of Pediatrics, Naomi Berrie Diabetes Center, Columbia University, New York, New York
| | - Ira J. Goldberg
- Department of Medicine, Preventive Medicine and Nutrition, Columbia University, New York, New York
| | - Yi-Hao Yu
- Department of Medicine, Preventive Medicine and Nutrition, Columbia University, New York, New York
- Corresponding author: Yi-Hao Yu,
| |
Collapse
|
28
|
Ruschke K, Ebelt H, Klöting N, Boettger T, Raum K, Blüher M, Braun T. Defective peripheral nerve development is linked to abnormal architecture and metabolic activity of adipose tissue in Nscl-2 mutant mice. PLoS One 2009; 4:e5516. [PMID: 19436734 PMCID: PMC2677458 DOI: 10.1371/journal.pone.0005516] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Accepted: 04/16/2009] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND In mammals the interplay between the peripheral nervous system (PNS) and adipose tissue is widely unexplored. We have employed mice, which develop an adult onset of obesity due to the lack the neuronal specific transcription factor Nscl-2 to investigate the interplay between the nervous system and white adipose tissue (WAT). METHODOLOGY Changes in the architecture and innervation of WAT were compared between wildtype, Nscl2-/-, ob/ob and Nscl2-/-//ob/ob mice using morphological methods, immunohistochemistry and flow cytometry. Metabolic alterations in mutant mice and in isolated cells were investigated under basal and stimulated conditions. PRINCIPAL FINDINGS We found that Nscl-2 mutant mice show a massive reduction of innervation of white epididymal and paired subcutaneous inguinal fat tissue including sensory and autonomic nerves as demonstrated by peripherin and neurofilament staining. Reduction of innervation went along with defects in the formation of the microvasculature, accumulation of cells of the macrophage/preadipocyte lineage, a bimodal distribution of the size of fat cells, and metabolic defects of isolated adipocytes. Despite a relative insulin resistance of white adipose tissue and isolated Nscl-2 mutant adipocytes the serum level of insulin in Nscl-2 mutant mice was only slightly increased. CONCLUSIONS We conclude that the reduction of the innervation and vascularization of WAT in Nscl-2 mutant mice leads to the increase of preadipocyte/macrophage-like cells, a bimodal distribution of the size of adipocytes in WAT and an altered metabolic activity of adipocytes.
Collapse
Affiliation(s)
- Karen Ruschke
- Institute of Physiological Chemistry, University of Halle-Wittenberg, Halle, Germany
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Henning Ebelt
- Institute of Physiological Chemistry, University of Halle-Wittenberg, Halle, Germany
| | - Nora Klöting
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Thomas Boettger
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Kay Raum
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Thomas Braun
- Institute of Physiological Chemistry, University of Halle-Wittenberg, Halle, Germany
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
29
|
Ahmadian M, Duncan RE, Varady KA, Frasson D, Hellerstein MK, Birkenfeld AL, Samuel VT, Shulman GI, Wang Y, Kang C, Sul HS. Adipose overexpression of desnutrin promotes fatty acid use and attenuates diet-induced obesity. Diabetes 2009; 58:855-66. [PMID: 19136649 PMCID: PMC2661591 DOI: 10.2337/db08-1644] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To investigate the role of desnutrin in adipose tissue triacylglycerol (TAG) and fatty acid metabolism. RESEARCH DESIGN AND METHODS We generated transgenic mice overexpressing desnutrin (also called adipose triglyceride lipase [ATGL]) in adipocytes (aP2-desnutrin) and also performed adenoviral-mediated overexpression of desnutrin in 3T3-L1CARDelta1 adipocytes. RESULTS aP2-desnutrin mice were leaner with decreased adipose tissue TAG content and smaller adipocyte size. Overexpression of desnutrin increased lipolysis but did not result in increased serum nonesterified fatty acid levels or ectopic TAG storage. We found increased cycling between diacylglycerol (DAG) and TAG and increased fatty acid oxidation in adipocytes from these mice, as well as improved insulin sensitivity. CONCLUSIONS We show that by increasing lipolysis, desnutrin overexpression causes reduced adipocyte TAG content and attenuation of diet-induced obesity. Desnutrin-mediated lipolysis promotes fatty acid oxidation and re-esterification within adipocytes.
Collapse
Affiliation(s)
- Maryam Ahmadian
- Department of Nutritional Science and Toxicology, University of California, Berkeley, California
| | - Robin E. Duncan
- Department of Nutritional Science and Toxicology, University of California, Berkeley, California
| | - Krista A. Varady
- Department of Nutritional Science and Toxicology, University of California, Berkeley, California
| | - Danubia Frasson
- Department of Nutritional Science and Toxicology, University of California, Berkeley, California
| | - Marc K. Hellerstein
- Department of Nutritional Science and Toxicology, University of California, Berkeley, California
| | - Andreas L. Birkenfeld
- Department of Internal Medicine, Yale University, School of Medicine, New Haven, Connecticut
| | - Varman T. Samuel
- Department of Internal Medicine, Yale University, School of Medicine, New Haven, Connecticut
| | - Gerald I. Shulman
- Department of Internal Medicine, Yale University, School of Medicine, New Haven, Connecticut
| | - Yuhui Wang
- Department of Nutritional Science and Toxicology, University of California, Berkeley, California
| | - Chulho Kang
- Department of Molecular and Cell Biology, University of California, Berkeley, California
| | - Hei Sook Sul
- Department of Nutritional Science and Toxicology, University of California, Berkeley, California
- Corresponding author: Hei Sook Sul,
| |
Collapse
|
30
|
Ryu D, Oh KJ, Jo HY, Hedrick S, Kim YN, Hwang YJ, Park TS, Han JS, Choi CS, Montminy M, Koo SH. TORC2 regulates hepatic insulin signaling via a mammalian phosphatidic acid phosphatase, LIPIN1. Cell Metab 2009; 9:240-51. [PMID: 19254569 DOI: 10.1016/j.cmet.2009.01.007] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2008] [Revised: 11/13/2008] [Accepted: 01/14/2009] [Indexed: 01/10/2023]
Abstract
TORC2 is a major transcriptional coactivator for hepatic glucose production. Insulin impedes gluconeogenesis by inhibiting TORC2 via SIK2-dependent phosphorylation at Ser171. Interruption of this process greatly perturbs hepatic glucose metabolism, thus promoting hyperglycemia in rodents. Here, we show that hyperactivation of TORC2 would exacerbate insulin resistance by enhancing expression of LIPIN1, a mammalian phosphatidic acid phosphatase for diacylglycerol (DAG) synthesis. Diet-induced or genetic obesity increases LIPIN1 expression in mouse liver, and TORC2 is responsible for its transcriptional activation. While overexpression of LIPIN1 disturbs hepatic insulin signaling, knockdown of LIPIN1 ameliorates hyperglycemia and insulin resistance by reducing DAG and PKCvarepsilon activity in db/db mice. Finally, TORC2-mediated insulin resistance is partially rescued by concomitant knockdown of LIPIN1, confirming the critical role of LIPIN1 in the perturbation of hepatic insulin signaling. These data propose that dysregulation of TORC2 would further exaggerate insulin resistance and promote type 2 diabetes in a LIPIN1-dependent manner.
Collapse
Affiliation(s)
- Dongryeol Ryu
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 300 Chunchun-dong, Jangan-gu, Suwon, Gyeonggi-do 440-746, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Nagai Y, Yonemitsu S, Erion DM, Iwasaki T, Stark R, Weismann D, Dong J, Zhang D, Jurczak MJ, Löffler MG, Cresswell J, Yu XX, Murray SF, Bhanot S, Monia BP, Bogan JS, Samuel V, Shulman GI. The role of peroxisome proliferator-activated receptor gamma coactivator-1 beta in the pathogenesis of fructose-induced insulin resistance. Cell Metab 2009; 9:252-64. [PMID: 19254570 PMCID: PMC3131094 DOI: 10.1016/j.cmet.2009.01.011] [Citation(s) in RCA: 158] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Revised: 12/12/2008] [Accepted: 01/29/2009] [Indexed: 10/21/2022]
Abstract
Peroxisome proliferator-activated receptor gamma coactivator-1 beta (PGC-1beta) is known to be a transcriptional coactivator for SREBP-1, the master regulator of hepatic lipogenesis. Here, we evaluated the role of PGC-1beta in the pathogenesis of fructose-induced insulin resistance by using an antisense oligonucletoide (ASO) to knockdown PGC-1beta in liver and adipose tissue. PGC-1beta ASO improved the metabolic phenotype induced by fructose feeding by reducing expression of SREBP-1 and downstream lipogenic genes in liver. PGC-1beta ASO also reversed hepatic insulin resistance induced by fructose in both basal and insulin-stimulated states. Furthermore, PGC-1beta ASO increased insulin-stimulated whole-body glucose disposal due to a threefold increase in glucose uptake in white adipose tissue. These data support an important role for PGC-1beta in the pathogenesis of fructose-induced insulin resistance and suggest that PGC-1beta inhibition may be a therapeutic target for treatment of NAFLD, hypertriglyceridemia, and insulin resistance associated with increased de novo lipogenesis.
Collapse
Affiliation(s)
- Yoshio Nagai
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06536-8012, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
AdPLA ablation increases lipolysis and prevents obesity induced by high-fat feeding or leptin deficiency. Nat Med 2009; 15:159-68. [PMID: 19136964 PMCID: PMC2863116 DOI: 10.1038/nm.1904] [Citation(s) in RCA: 202] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Accepted: 11/19/2008] [Indexed: 01/04/2023]
Abstract
A main function of white adipose tissue is to release fatty acids from triacylglycerol for other tissues to use as an energy source. While endocrine regulation of lipolysis has been extensively studied, autocrine/paracrine regulation is not well understood. Here, we describe the role of AdPLA, the newly identified major adipocyte phospholipase A2, in the regulation of lipolysis and adiposity. AdPLA null mice have a markedly higher rate of lipolysis, due to increased cAMP levels arising from the marked reduction in adipose PGE2 that binds the Gαi-coupled receptor, EP3. AdPLA null mice have drastically reduced adipose tissue mass and triglyceride content, with normal adipogenesis. They also have higher energy expenditure with higher fatty acid oxidation within adipocytes. AdPLA deficient ob/ob mice remain hyperphagic but lean, with increased energy expenditure, yet have ectopic triglyceride storage and insulin resistance. AdPLA is a major regulator of adipocyte lipolysis and critical for the development of obesity.
Collapse
|
33
|
Essop MF, Camp HS, Choi CS, Sharma S, Fryer RM, Reinhart GA, Guthrie PH, Bentebibel A, Gu Z, Shulman GI, Taegtmeyer H, Wakil SJ, Abu-Elheiga L. Reduced heart size and increased myocardial fuel substrate oxidation in ACC2 mutant mice. Am J Physiol Heart Circ Physiol 2008; 295:H256-65. [PMID: 18487439 DOI: 10.1152/ajpheart.91489.2007] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The cardiac-enriched isoform of acetyl-CoA carboxylase (ACC2) is a key regulator of mitochondrial fatty acid (FA) uptake via carnitine palmitoyltransferase 1 (CPT1). To test the hypothesis that oxidative metabolism is upregulated in hearts from animals lacking ACC2 (employing a transgenic Acc2-mutant mouse), we assessed cardiac function in vivo and determined rates of myocardial substrate oxidation ex vivo. When examined by echocardiography, there was no difference in systolic function, but left ventricular mass of the Acc2-mutant (MUT) mouse was significantly reduced ( approximately 25%) compared with wild-types (WT). Reduced activation of the mammalian target of rapamycin (mTOR) and its downstream target p70S6K was found in MUT hearts. Exogenous oxidation rates of oleate were increased approximately 22%, and, unexpectedly, exogenous glucose oxidation rates were also increased in MUT hearts. Using a hyperinsulinemic-euglycemic clamp, we found that glucose uptake in MUT hearts was increased by approximately 83%. Myocardial triglyceride levels were significantly reduced in MUT vs. WT while glycogen content was the same. In parallel, transcript levels of PPARalpha and its target genes, pyruvate dehydrogenase kinase-4 (PDK-4), malonyl-CoA decarboxylase (MCD), and mCPT1, were downregulated in MUT mice. In summary, we report that 1) Acc2-mutant hearts exhibit a marked preference for the oxidation of both glucose and FAs coupled with greater utilization of endogenous fuel substrates (triglycerides), 2) attenuated mTOR signaling may result in reduced heart sizes observed in Acc2-mutant mice, and 3) Acc2-mutant hearts displayed normal functional parameters despite a significant decrease in size.
Collapse
Affiliation(s)
- M Faadiel Essop
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Continuous fat oxidation in acetyl-CoA carboxylase 2 knockout mice increases total energy expenditure, reduces fat mass, and improves insulin sensitivity. Proc Natl Acad Sci U S A 2007; 104:16480-5. [PMID: 17923673 DOI: 10.1073/pnas.0706794104] [Citation(s) in RCA: 244] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Acetyl-CoA carboxylase 2 (ACC)2 is a key regulator of mitochondrial fat oxidation. To examine the impact of ACC2 deletion on whole-body energy metabolism, we measured changes in substrate oxidation and total energy expenditure in Acc2(-/-) and WT control mice fed either regular or high-fat diets. To determine insulin action in vivo, we also measured whole-body insulin-stimulated liver and muscle glucose metabolism during a hyperinsulinemic-euglycemic clamp in Acc2(-/-) and WT control mice fed a high-fat diet. Contrary to previous studies that have suggested that increased fat oxidation might result in lower glucose oxidation, both fat and carbohydrate oxidation were simultaneously increased in Acc2(-/-) mice. This increase in both fat and carbohydrate oxidation resulted in an increase in total energy expenditure, reductions in fat and lean body mass and prevention from diet-induced obesity. Furthermore, Acc2(-/-) mice were protected from fat-induced peripheral and hepatic insulin resistance. These improvements in insulin-stimulated glucose metabolism were associated with reduced diacylglycerol content in muscle and liver, decreased PKC activity in muscle and PKCepsilon activity in liver, and increased insulin-stimulated Akt2 activity in these tissues. Taken together with previous work demonstrating that Acc2(-/-) mice have a normal lifespan, these data suggest that Acc2 inhibition is a viable therapeutic option for the treatment of obesity and type 2 diabetes.
Collapse
|
35
|
Farese RV, Sajan MP, Yang H, Li P, Mastorides S, Gower WR, Nimal S, Choi CS, Kim S, Shulman GI, Kahn CR, Braun U, Leitges M. Muscle-specific knockout of PKC-lambda impairs glucose transport and induces metabolic and diabetic syndromes. J Clin Invest 2007. [PMID: 17641777 DOI: 10.1172/jci31408c1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Obesity, the metabolic syndrome, and type 2 diabetes mellitus (T2DM) are major global health problems. Insulin resistance is frequently present in these disorders, but the causes and effects of such resistance are unknown. Here, we generated mice with muscle-specific knockout of the major murine atypical PKC (aPKC), PKC-lambda, a postulated mediator for insulin-stimulated glucose transport. Glucose transport and translocation of glucose transporter 4 (GLUT4) to the plasma membrane were diminished in muscles of both homozygous and heterozygous PKC-lambda knockout mice and were accompanied by systemic insulin resistance; impaired glucose tolerance or diabetes; islet beta cell hyperplasia; abdominal adiposity; hepatosteatosis; elevated serum triglycerides, FFAs, and LDL-cholesterol; and diminished HDL-cholesterol. In contrast to the defective activation of muscle aPKC, insulin signaling and actions were intact in muscle, liver, and adipocytes. These findings demonstrate the importance of aPKC in insulin-stimulated glucose transport in muscles of intact mice and show that insulin resistance and resultant hyperinsulinemia owing to a specific defect in muscle aPKC is sufficient to induce abdominal obesity and other lipid abnormalities of the metabolic syndrome and T2DM. These findings are particularly relevant because humans who have obesity, impaired glucose tolerance, and T2DM reportedly have defective activation and/or diminished levels of muscle aPKC.
Collapse
Affiliation(s)
- Robert V Farese
- James A. Haley Veterans Medical Center, Tampa, Florida 33612, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Farese RV, Sajan MP, Yang H, Li P, Mastorides S, Gower WR, Nimal S, Choi CS, Kim S, Shulman GI, Kahn CR, Braun U, Leitges M. Muscle-specific knockout of PKC-lambda impairs glucose transport and induces metabolic and diabetic syndromes. J Clin Invest 2007; 117:2289-301. [PMID: 17641777 PMCID: PMC1913489 DOI: 10.1172/jci31408] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2007] [Accepted: 05/08/2007] [Indexed: 12/11/2022] Open
Abstract
Obesity, the metabolic syndrome, and type 2 diabetes mellitus (T2DM) are major global health problems. Insulin resistance is frequently present in these disorders, but the causes and effects of such resistance are unknown. Here, we generated mice with muscle-specific knockout of the major murine atypical PKC (aPKC), PKC-lambda, a postulated mediator for insulin-stimulated glucose transport. Glucose transport and translocation of glucose transporter 4 (GLUT4) to the plasma membrane were diminished in muscles of both homozygous and heterozygous PKC-lambda knockout mice and were accompanied by systemic insulin resistance; impaired glucose tolerance or diabetes; islet beta cell hyperplasia; abdominal adiposity; hepatosteatosis; elevated serum triglycerides, FFAs, and LDL-cholesterol; and diminished HDL-cholesterol. In contrast to the defective activation of muscle aPKC, insulin signaling and actions were intact in muscle, liver, and adipocytes. These findings demonstrate the importance of aPKC in insulin-stimulated glucose transport in muscles of intact mice and show that insulin resistance and resultant hyperinsulinemia owing to a specific defect in muscle aPKC is sufficient to induce abdominal obesity and other lipid abnormalities of the metabolic syndrome and T2DM. These findings are particularly relevant because humans who have obesity, impaired glucose tolerance, and T2DM reportedly have defective activation and/or diminished levels of muscle aPKC.
Collapse
Affiliation(s)
- Robert V Farese
- James A. Haley Veterans Medical Center, Tampa, Florida 33612, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Choi CS, Fillmore JJ, Kim JK, Liu ZX, Kim S, Collier EF, Kulkarni A, Distefano A, Hwang YJ, Kahn M, Chen Y, Yu C, Moore IK, Reznick RM, Higashimori T, Shulman GI. Overexpression of uncoupling protein 3 in skeletal muscle protects against fat-induced insulin resistance. J Clin Invest 2007; 117:1995-2003. [PMID: 17571165 PMCID: PMC1888566 DOI: 10.1172/jci13579] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2001] [Accepted: 04/24/2007] [Indexed: 01/04/2023] Open
Abstract
Insulin resistance is a major factor in the pathogenesis of type 2 diabetes and is strongly associated with obesity. Increased concentrations of intracellular fatty acid metabolites have been postulated to interfere with insulin signaling by activation of a serine kinase cascade involving PKCtheta in skeletal muscle. Uncoupling protein 3 (UCP3) has been postulated to dissipate the mitochondrial proton gradient and cause metabolic inefficiency. We therefore hypothesized that overexpression of UCP3 in skeletal muscle might protect against fat-induced insulin resistance in muscle by conversion of intramyocellular fat into thermal energy. Wild-type mice fed a high-fat diet were markedly insulin resistant, a result of defects in insulin-stimulated glucose uptake in skeletal muscle and hepatic insulin resistance. Insulin resistance in these tissues was associated with reduced insulin-stimulated insulin receptor substrate 1- (IRS-1-) and IRS-2-associated PI3K activity in muscle and liver, respectively. In contrast, UCP3-overexpressing mice were completely protected against fat-induced defects in insulin signaling and action in these tissues. Furthermore, these changes were associated with a lower membrane-to-cytosolic ratio of diacylglycerol and reduced PKCtheta activity in whole-body fat-matched UCP3 transgenic mice. These results suggest that increasing mitochondrial uncoupling in skeletal muscle may be an excellent therapeutic target for type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Cheol Soo Choi
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jonathan J. Fillmore
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jason K. Kim
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zhen-Xiang Liu
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sheene Kim
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emily F. Collier
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ameya Kulkarni
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Alberto Distefano
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yu-Jin Hwang
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Mario Kahn
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yan Chen
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Chunli Yu
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Irene K. Moore
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Richard M. Reznick
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Takamasa Higashimori
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Gerald I. Shulman
- Department of Internal Medicine,
Department of Cellular and Molecular Physiology, and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
38
|
Choi CS, Savage DB, Kulkarni A, Yu XX, Liu ZX, Morino K, Kim S, Distefano A, Samuel VT, Neschen S, Zhang D, Wang A, Zhang XM, Kahn M, Cline GW, Pandey SK, Geisler JG, Bhanot S, Monia BP, Shulman GI. Suppression of diacylglycerol acyltransferase-2 (DGAT2), but not DGAT1, with antisense oligonucleotides reverses diet-induced hepatic steatosis and insulin resistance. J Biol Chem 2007; 282:22678-88. [PMID: 17526931 DOI: 10.1074/jbc.m704213200] [Citation(s) in RCA: 294] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major contributing factor to hepatic insulin resistance in type 2 diabetes. Diacylglycerol acyltransferase (Dgat), of which there are two isoforms (Dgat1 and Dgat2), catalyzes the final step in triglyceride synthesis. We evaluated the metabolic impact of pharmacological reduction of DGAT1 and -2 expression in liver and fat using antisense oligonucleotides (ASOs) in rats with diet-induced NAFLD. Dgat1 and Dgat2 ASO treatment selectively reduced DGAT1 and DGAT2 mRNA levels in liver and fat, but only Dgat2 ASO treatment significantly reduced hepatic lipids (diacylglycerol and triglyceride but not long chain acyl CoAs) and improved hepatic insulin sensitivity. Because Dgat catalyzes triglyceride synthesis from diacylglycerol, and because we have hypothesized that diacylglycerol accumulation triggers fat-induced hepatic insulin resistance through protein kinase C epsilon activation, we next sought to understand the paradoxical reduction in diacylglycerol in Dgat2 ASO-treated rats. Within 3 days of starting Dgat2 ASO therapy in high fat-fed rats, plasma fatty acids increased, whereas hepatic lysophosphatidic acid and diacylglycerol levels were similar to those of control rats. These changes were associated with reduced expression of lipogenic genes (SREBP1c, ACC1, SCD1, and mtGPAT) and increased expression of oxidative/thermogenic genes (CPT1 and UCP2). Taken together, these data suggest that knocking down Dgat2 protects against fat-induced hepatic insulin resistance by paradoxically lowering hepatic diacylglycerol content and protein kinase C epsilon activation through decreased SREBP1c-mediated lipogenesis and increased hepatic fatty acid oxidation.
Collapse
Affiliation(s)
- Cheol Soo Choi
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Samuel VT, Liu ZX, Wang A, Beddow SA, Geisler JG, Kahn M, Zhang XM, Monia BP, Bhanot S, Shulman GI. Inhibition of protein kinase Cepsilon prevents hepatic insulin resistance in nonalcoholic fatty liver disease. J Clin Invest 2007; 117:739-45. [PMID: 17318260 PMCID: PMC1797607 DOI: 10.1172/jci30400] [Citation(s) in RCA: 380] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2006] [Accepted: 01/03/2007] [Indexed: 12/25/2022] Open
Abstract
Nonalcoholic fatty liver disease is strongly associated with hepatic insulin resistance and type 2 diabetes mellitus, but the molecular signals linking hepatic fat accumulation to hepatic insulin resistance are unknown. Three days of high-fat feeding in rats results specifically in hepatic steatosis and hepatic insulin resistance. In this setting, PKCepsilon, but not other isoforms of PKC, is activated. To determine whether PKCepsilon plays a causal role in the pathogenesis of hepatic insulin resistance, we treated rats with an antisense oligonucleotide against PKCepsilon and subjected them to 3 days of high-fat feeding. Knocking down PKCepsilon expression protects rats from fat-induced hepatic insulin resistance and reverses fat-induced defects in hepatic insulin signaling. Furthermore, we show that PKCepsilon associates with the insulin receptor in vivo and impairs insulin receptor kinase activity both in vivo and in vitro. These data support the hypothesis that PKCepsilon plays a critical role in mediating fat-induced hepatic insulin resistance and represents a novel therapeutic target for type 2 diabetes.
Collapse
Affiliation(s)
- Varman T. Samuel
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Veterans Administration Medical Center, West Haven, Connecticut, USA.
Isis Pharmaceuticals Inc., Carlsbad, California, USA.
Department of Cellular and Molecular Physiology and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zhen-Xiang Liu
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Veterans Administration Medical Center, West Haven, Connecticut, USA.
Isis Pharmaceuticals Inc., Carlsbad, California, USA.
Department of Cellular and Molecular Physiology and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Amy Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Veterans Administration Medical Center, West Haven, Connecticut, USA.
Isis Pharmaceuticals Inc., Carlsbad, California, USA.
Department of Cellular and Molecular Physiology and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sara A. Beddow
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Veterans Administration Medical Center, West Haven, Connecticut, USA.
Isis Pharmaceuticals Inc., Carlsbad, California, USA.
Department of Cellular and Molecular Physiology and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - John G. Geisler
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Veterans Administration Medical Center, West Haven, Connecticut, USA.
Isis Pharmaceuticals Inc., Carlsbad, California, USA.
Department of Cellular and Molecular Physiology and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Mario Kahn
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Veterans Administration Medical Center, West Haven, Connecticut, USA.
Isis Pharmaceuticals Inc., Carlsbad, California, USA.
Department of Cellular and Molecular Physiology and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Xian-man Zhang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Veterans Administration Medical Center, West Haven, Connecticut, USA.
Isis Pharmaceuticals Inc., Carlsbad, California, USA.
Department of Cellular and Molecular Physiology and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Brett P. Monia
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Veterans Administration Medical Center, West Haven, Connecticut, USA.
Isis Pharmaceuticals Inc., Carlsbad, California, USA.
Department of Cellular and Molecular Physiology and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sanjay Bhanot
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Veterans Administration Medical Center, West Haven, Connecticut, USA.
Isis Pharmaceuticals Inc., Carlsbad, California, USA.
Department of Cellular and Molecular Physiology and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Gerald I. Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Veterans Administration Medical Center, West Haven, Connecticut, USA.
Isis Pharmaceuticals Inc., Carlsbad, California, USA.
Department of Cellular and Molecular Physiology and
Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
40
|
Samuel VT, Liu ZX, Wang A, Beddow SA, Geisler JG, Kahn M, Zhang XM, Monia BP, Bhanot S, Shulman GI. Inhibition of protein kinase Cepsilon prevents hepatic insulin resistance in nonalcoholic fatty liver disease. J Clin Invest 2007. [PMID: 17318260 DOI: 10.1172/jci3040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Nonalcoholic fatty liver disease is strongly associated with hepatic insulin resistance and type 2 diabetes mellitus, but the molecular signals linking hepatic fat accumulation to hepatic insulin resistance are unknown. Three days of high-fat feeding in rats results specifically in hepatic steatosis and hepatic insulin resistance. In this setting, PKCepsilon, but not other isoforms of PKC, is activated. To determine whether PKCepsilon plays a causal role in the pathogenesis of hepatic insulin resistance, we treated rats with an antisense oligonucleotide against PKCepsilon and subjected them to 3 days of high-fat feeding. Knocking down PKCepsilon expression protects rats from fat-induced hepatic insulin resistance and reverses fat-induced defects in hepatic insulin signaling. Furthermore, we show that PKCepsilon associates with the insulin receptor in vivo and impairs insulin receptor kinase activity both in vivo and in vitro. These data support the hypothesis that PKCepsilon plays a critical role in mediating fat-induced hepatic insulin resistance and represents a novel therapeutic target for type 2 diabetes.
Collapse
Affiliation(s)
- Varman T Samuel
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06536, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Samuel VT, Liu ZX, Wang A, Beddow SA, Geisler JG, Kahn M, Zhang XM, Monia BP, Bhanot S, Shulman GI. Inhibition of protein kinase Cepsilon prevents hepatic insulin resistance in nonalcoholic fatty liver disease. J Clin Invest 2007. [PMID: 17318260 DOI: 10.1172/jci130400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Nonalcoholic fatty liver disease is strongly associated with hepatic insulin resistance and type 2 diabetes mellitus, but the molecular signals linking hepatic fat accumulation to hepatic insulin resistance are unknown. Three days of high-fat feeding in rats results specifically in hepatic steatosis and hepatic insulin resistance. In this setting, PKCepsilon, but not other isoforms of PKC, is activated. To determine whether PKCepsilon plays a causal role in the pathogenesis of hepatic insulin resistance, we treated rats with an antisense oligonucleotide against PKCepsilon and subjected them to 3 days of high-fat feeding. Knocking down PKCepsilon expression protects rats from fat-induced hepatic insulin resistance and reverses fat-induced defects in hepatic insulin signaling. Furthermore, we show that PKCepsilon associates with the insulin receptor in vivo and impairs insulin receptor kinase activity both in vivo and in vitro. These data support the hypothesis that PKCepsilon plays a critical role in mediating fat-induced hepatic insulin resistance and represents a novel therapeutic target for type 2 diabetes.
Collapse
Affiliation(s)
- Varman T Samuel
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06536, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Vianna CR, Huntgeburth M, Coppari R, Choi CS, Lin J, Krauss S, Barbatelli G, Tzameli I, Kim YB, Cinti S, Shulman GI, Spiegelman BM, Lowell BB. Hypomorphic mutation of PGC-1beta causes mitochondrial dysfunction and liver insulin resistance. Cell Metab 2006; 4:453-64. [PMID: 17141629 PMCID: PMC1764615 DOI: 10.1016/j.cmet.2006.11.003] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2006] [Revised: 10/03/2006] [Accepted: 11/06/2006] [Indexed: 01/06/2023]
Abstract
PGC-1beta is a transcriptional coactivator that potently stimulates mitochondrial biogenesis and respiration of cells. Here, we have generated mice lacking exons 3 to 4 of the Pgc-1beta gene (Pgc-1beta(E3,4-/E3,4-) mice). These mice express a mutant protein that has reduced coactivation activity on a subset of transcription factors, including ERRalpha, a major target of PGC-1beta in the induction of mitochondrial gene expression. The mutant mice have reduced expression of OXPHOS genes and mitochondrial dysfunction in liver and skeletal muscle as well as elevated liver triglycerides. Euglycemic-hyperinsulinemic clamp and insulin signaling studies show that PGC-1beta mutant mice have normal skeletal muscle response to insulin but have hepatic insulin resistance. These results demonstrate that PGC-1beta is required for normal expression of OXPHOS genes and mitochondrial function in liver and skeletal muscle. Importantly, these abnormalities do not cause insulin resistance in skeletal muscle but cause substantially reduced insulin action in the liver.
Collapse
MESH Headings
- Animals
- Gene Expression Regulation/drug effects
- Glucose Clamp Technique
- Hypoglycemic Agents/pharmacology
- Insulin/pharmacology
- Insulin Resistance/genetics
- Liver/metabolism
- Liver/pathology
- Mice
- Mice, Knockout
- Mitochondria, Liver/genetics
- Mitochondria, Liver/metabolism
- Mitochondria, Liver/pathology
- Mitochondria, Muscle/genetics
- Mitochondria, Muscle/metabolism
- Mitochondria, Muscle/pathology
- Mitochondrial Proteins/biosynthesis
- Mitochondrial Proteins/genetics
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Mutation
- Organ Specificity
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Trans-Activators/deficiency
- Trans-Activators/metabolism
- Transcription Factors
- ERRalpha Estrogen-Related Receptor
Collapse
Affiliation(s)
- Claudia R. Vianna
- Department of Medicine, Division of Endocrinology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Michael Huntgeburth
- Department of Medicine, Division of Endocrinology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
- Clinic III for Internal Medicine, University of Cologne, 50937 Cologne, Germany
| | - Roberto Coppari
- Department of Medicine, Division of Endocrinology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Cheol Soo Choi
- Department of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Jiandie Lin
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Stefan Krauss
- Department of Medicine, Division of Endocrinology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Giorgio Barbatelli
- Institute of Normal Human Morphology, Faculty of Medicine, University of Marche, Ancona 60020, Italy
| | - Iphigenia Tzameli
- Department of Medicine, Division of Endocrinology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Young-Bum Kim
- Department of Medicine, Division of Endocrinology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Saverio Cinti
- Institute of Normal Human Morphology, Faculty of Medicine, University of Marche, Ancona 60020, Italy
| | - Gerald I. Shulman
- Department of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Bruce M. Spiegelman
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Bradford B. Lowell
- Department of Medicine, Division of Endocrinology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| |
Collapse
|
43
|
Xu D, Dhillon AS, Davey CG, Fournier PA, Palmer TN. Alcohol and glucose metabolism in skeletal muscles in the rat. Addict Biol 2003; 1:71-83. [PMID: 12893488 DOI: 10.1080/1355621961000124706] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Ethanol is known to cause an acute and profound insulin resistance in man and the rat primarily via effects on glucose utilization. This paper examines the nature of these inhibitory effects on whole-body glucose utilization using the euglycaemic hyperinsulinaemic clamp in the conscious unrestrained rat. We confirm that ethanol infusion causes an acute insulin resistance, the rate of glucose infusion required to maintain euglycaemia (GIR) being decreased markedly by ethanol. To ensure that the GIR is a measure of whole-body glucose disposal, glucose turnover and hepatic glycogen levels were measured. These studies showed that ethanol totally suppressed hepatic glucose production. The reduction in GIR is associated with marked decreases in glucose uptake and glycogen synthesis in most skeletal muscles. In oxidative but not in non-oxidative muscles, the activation of glycogen synthase in response to insulin was decreased by ethanol, suggesting that a defect in glycogen synthase activation may be responsible for the decrease in glycogen synthesis. The basis of the inhibitory effects of ethanol on insulin-stimulated glucose metabolism in muscle is unknown, but may involve membrane-associated impairments in insulin signalling and/or the glucose transport system.
Collapse
Affiliation(s)
- D Xu
- Department of Biochemistry, University of Western Australia, Nedlands, Australia
| | | | | | | | | |
Collapse
|
44
|
Abstract
The purpose of this review is to summarize our current understanding of the acute and chronic interactions between alcohol and nutrient metabolism in skeletal muscle. Insulin is well known to play an important regulatory role in nutrient, especially glucose, uptake and utilization in skeletal muscle. Several studies have shown that alcohol can acutely reduce the normal metabolic responses of skeletal muscle to the action of insulin. The most obvious of these is an acute impairment in glucose metabolism associated with alcohol consumption. While the exact mechanism(s) underlying this acute insulin resistance is presently unclear, several possible factors are discussed in this review. In contrast to these short-term effects, the effects of alcohol on skeletal muscle insulin sensitivity in chronic alcohol abusers are not as well established. Chronic alcohol abuse is known to be associated with skeletal myopathies, believed to result from alcohol induced abnormalities in muscle protein synthesis. Finally, the alcohol-mediated impairments of many aspects of skeletal muscle metabolism are discussed in relation to the insulin resistance associated broad spectrum of common lifestyle-related disorders, including non-insulin dependent diabetes mellitus and obesity, the consequences of which may be important to the pathogenesis of alcohol-related diseases.
Collapse
Affiliation(s)
- D Xu
- Department of Biochemistry, University of Western Australia, Nedlands, Western Australia 6907, Australia
| | | | | |
Collapse
|
45
|
McCutcheon LJ, Geor RJ, Hinchcliff KW. Changes in skeletal muscle GLUT4 content and muscle membrane glucose transport following 6 weeks of exercise training. Equine Vet J 2002:199-204. [PMID: 12405686 DOI: 10.1111/j.2042-3306.2002.tb05418.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
This study examined changes in skeletal muscle GLUT4 content and glucose transport in isolated muscle membranes (GT) from horses before and 2 min after standardised submaximal exercise tests (SET) prior to and after completion of 6 weeks of training. Seven horses, age 3-9 years, body mass mean +/- s.e. 530 +/- 19 kg, and sedentary for at least 4 months, completed 6 weeks of training on a treadmill. An initial SET (UT) was performed on a 4 degree incline at a speed equivalent to 55% of pretraining VO2max and was repeated post-training at the same absolute workload (ABS). A third SET (REL) was performed at 55% of post-training VO2max. There was no significant pre- to postexercise change in GLUT4 content before or after training. Following training, total GLUT4 content was increased 2- or 3-fold in pre-exercise biopsies (pre UT: 0.30 +/- 0.05; pre ABS: 1.05 +/- 0.32; pre REL: 1.34 +/- 0.28 arbitrary units) (P<0.05) with similar increases in postexercise GLUT4 content (P<0.05) (post UT: 033 +/- 0.06; post ABS: 1.19 +/- 0.44; post REL: 1.43 +/- 0.31). GT increased 2.5- to 6-fold in postexercise muscle membrane vesicles in UT over a range of glucose concentrations. After training (ABS and REL), there was a 25-50% attenuation (P<0.05) in membrane GT in response to exercise in ABS and REL. These finding indicate that moderate intensity exercise training increased middle gluteal muscle GLUT4 content, but this change was not reflected in an increase in muscle membrane glucose transport activity in postexercise muscle samples.
Collapse
Affiliation(s)
- L J McCutcheon
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Canada
| | | | | |
Collapse
|
46
|
Kim JK, Zisman A, Fillmore JJ, Peroni OD, Kotani K, Perret P, Zong H, Dong J, Kahn CR, Kahn BB, Shulman GI. Glucose toxicity and the development of diabetes in mice with muscle-specific inactivation of GLUT4. J Clin Invest 2001; 108:153-60. [PMID: 11435467 PMCID: PMC353719 DOI: 10.1172/jci10294] [Citation(s) in RCA: 144] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Using cre/loxP gene targeting, transgenic mice with muscle-specific inactivation of the GLUT4 gene (muscle GLUT4 KO) were generated and shown to develop a diabetes phenotype. To determine the mechanism, we examined insulin-stimulated glucose uptake and metabolism during hyperinsulinemic-euglycemic clamp in control and muscle GLUT4 KO mice before and after development of diabetes. Insulin-stimulated whole body glucose uptake was decreased by 55% in muscle GLUT4 KO mice, an effect that could be attributed to a 92% decrease in insulin-stimulated muscle glucose uptake. Surprisingly, insulin's ability to stimulate adipose tissue glucose uptake and suppress hepatic glucose production was significantly impaired in muscle GLUT4 KO mice. To address whether these latter changes were caused by glucose toxicity, we treated muscle GLUT4 KO mice with phloridzin to prevent hyperglycemia and found that insulin-stimulated whole body and skeletal muscle glucose uptake were decreased substantially, whereas insulin-stimulated glucose uptake in adipose tissue and suppression of hepatic glucose production were normal after phloridzin treatment. In conclusion, these findings demonstrate that a primary defect in muscle glucose transport can lead to secondary defects in insulin action in adipose tissue and liver due to glucose toxicity. These secondary defects contribute to insulin resistance and to the development of diabetes.
Collapse
Affiliation(s)
- J K Kim
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06536-8012, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Casalino-Matsuda SM, Linares JA, Campos LM, Goldraij A. Influence of insulin on the metabolism of glucose in uteri isolated from ovariectomized and non ovariectomized underfed rats. Prostaglandins Leukot Essent Fatty Acids 2000; 63:337-41. [PMID: 11133170 DOI: 10.1054/plef.2000.0224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The effects of insulin on the metabolism of U14C-glucose in uteri isolated from ovariectomized and non-ovariectomized rats receiving a restricted diet (50% of the normal food intake) for 25 days, were studied. As a result of food restriction, the production of 14CO2 diminishes in intact rats, while results are reversed in ovariectomized ones. Various concentrations of insulin were added to the medium, but only 0.50 IU. ml(-1)was effective in increasing glucose metabolism in intact rats receiving a restricted diet; neither underfed castrated animals nor control ones receiving a normal diet, reacted to this concentration. The increase of 14CO2 produced by insulin is not affected by acetyl salicylic acid. Insulin does not alter the effect of underfeeding over arachidonic acid metabolism. On the contrary, the increase in glucose metabolism was blocked by N(G)methyl-L-arginine or by hemoglobin, increased with the addition of L arginine and is not affected by acetyl salicylic acid. Hemoglobin and L-arginine show no effects without insulin. We can conclude that the stimulating effect of insulin on glucose metabolism in uteri isolated from intact rats subjected to dietary restriction, is nitric oxide dependent.
Collapse
|
48
|
Kim JK, Michael MD, Previs SF, Peroni OD, Mauvais-Jarvis F, Neschen S, Kahn BB, Kahn CR, Shulman GI. Redistribution of substrates to adipose tissue promotes obesity in mice with selective insulin resistance in muscle. J Clin Invest 2000; 105:1791-7. [PMID: 10862794 PMCID: PMC378504 DOI: 10.1172/jci8305] [Citation(s) in RCA: 251] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Obesity and insulin resistance in skeletal muscle are two major factors in the pathogenesis of type 2 diabetes. Mice with muscle-specific inactivation of the insulin receptor gene (MIRKO) are normoglycemic but have increased fat mass. To identify the potential mechanism for this important association, we examined insulin action in specific tissues of MIRKO and control mice under hyperinsulinemic-euglycemic conditions. We found that insulin-stimulated muscle glucose transport and glycogen synthesis were decreased by about 80% in MIRKO mice, whereas insulin-stimulated fat glucose transport was increased threefold in MIRKO mice. These data demonstrate that selective insulin resistance in muscle promotes redistribution of substrates to adipose tissue thereby contributing to increased adiposity and development of the prediabetic syndrome.
Collapse
Affiliation(s)
- J K Kim
- Department of Internal Medicine and the Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Fery F, Plat L, Balasse EO. Effect of fasting on the intracellular metabolic partition of intravenously infused glucose in humans. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:E815-23. [PMID: 10567007 DOI: 10.1152/ajpendo.1999.277.5.e815] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effects of fasting on the pathways of insulin-stimulated glucose disposal were explored in three groups of seven normal subjects. Group 1 was submitted to a euglycemic hyperinsulinemic clamp ( approximately 100 microU/ml) after both a 12-h and a 4-day fast. The combined use of [3-(3)H]- and [U-(14)C]glucose allowed us to demonstrate that fasting inhibits, by approximately 50%, glucose disposal, glycolysis, glucose oxidation, and glycogen synthesis via the direct pathway. In group 2, in which the clamp glucose disposal during fasting was restored by hyperglycemia (155 +/- 15 mg/dl), fasting stimulated glycogen synthesis (+29 +/- 2%) and inhibited glycolysis (-32 +/- 3%) but only in its oxidative component (-40 +/- 3%). Results were similar in group 3 in which the clamp glucose disposal was restored by a pharmacological elevation of insulin ( approximately 2,800 microU/ml), but in this case, both glycogen synthesis and nonoxidative glycolysis participated in the rise in nonoxidative glucose disposal. In all groups, the reduction in total carbohydrate oxidation (indirect calorimetry) induced by fasting markedly exceeded the reduction in circulating glucose oxidation, suggesting that fasting also inhibits intracellular glycogen oxidation. Thus prior fasting favors glycogen retention by three mechanisms: 1) stimulation of glycogen synthesis via the direct pathway; 2) preferential inhibition of oxidative rather than nonoxidative glycolysis, thus allowing carbon conservation for glycogen synthesis via the indirect pathway; and 3) suppression of intracellular glycogen oxidation.
Collapse
Affiliation(s)
- F Fery
- Laboratory of Experimental Medicine, Department of Endocrinology, Erasmus Hospital, University of Brussels, B-1070 Brussels, Belgium
| | | | | |
Collapse
|
50
|
Kim JK, Choi CS, Youn JH. Acute effect of growth hormone to induce peripheral insulin resistance is independent of FFA and insulin levels in rats. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:E742-9. [PMID: 10516135 DOI: 10.1152/ajpendo.1999.277.4.e742] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To examine whether growth hormone (GH) induces peripheral insulin resistance by altering plasma free fatty acid (FFA) or insulin levels, the effects of GH infusion on insulin-stimulated glucose fluxes were studied in conscious rats under two protocols. In study 1, either saline (n = 7) or human recombinant GH (21 microg. kg(-1). h(-1); n = 8) was infused for 300 min, and insulin-stimulated glucose fluxes were estimated during the final 150-min period of hyperinsulinemic euglycemic clamps. In study 2, hyperinsulinemic euglycemic clamps were first conducted for 150 min (to raise plasma insulin and suppress FFA levels), and saline or GH (n = 7 for each) was subsequently infused for the following 300-min clamp period. In study 1, GH infusion in the basal state did not significantly alter plasma FFA or insulin levels. In contrast, GH infusion decreased insulin-stimulated glucose uptake, glycolysis, and glycogen synthesis by 32, 27, and 40%, respectively (P < 0.05). In study 2, GH infusion during hyperinsulinemic euglycemic clamps did not alter plasma FFA or insulin levels (P > 0.05). GH infusion had no effect on insulin-stimulated glucose uptake during the initial 150 min but eventually decreased insulin-stimulated glucose uptake by 37% (P < 0. 05), similar to the results in study 1. These data indicate that GH induces peripheral insulin resistance independent of plasma FFA and insulin levels. The induction of insulin resistance was preceded by suppression of glycogen synthesis, consistent with the hypothesis that metabolic impairment precedes and causes development of peripheral insulin resistance.
Collapse
Affiliation(s)
- J K Kim
- Department of Physiology, University of Southern California School of Medicine, Los Angeles, California 90089-9142, USA
| | | | | |
Collapse
|