151
|
Neinast MD, Jang C, Hui S, Murashige DS, Chu Q, Morscher RJ, Li X, Zhan L, White E, Anthony TG, Rabinowitz JD, Arany Z. Quantitative Analysis of the Whole-Body Metabolic Fate of Branched-Chain Amino Acids. Cell Metab 2019; 29:417-429.e4. [PMID: 30449684 PMCID: PMC6365191 DOI: 10.1016/j.cmet.2018.10.013] [Citation(s) in RCA: 335] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 05/25/2018] [Accepted: 10/22/2018] [Indexed: 01/04/2023]
Abstract
Elevations in branched-chain amino acids (BCAAs) associate with numerous systemic diseases, including cancer, diabetes, and heart failure. However, an integrated understanding of whole-body BCAA metabolism remains lacking. Here, we employ in vivo isotopic tracing to systemically quantify BCAA oxidation in healthy and insulin-resistant mice. We find that most tissues rapidly oxidize BCAAs into the tricarboxylic acid (TCA) cycle, with the greatest quantity occurring in muscle, brown fat, liver, kidneys, and heart. Notably, pancreas supplies 20% of its TCA carbons from BCAAs. Genetic and pharmacologic suppression of branched-chain alpha-ketoacid dehydrogenase kinase, a clinically targeted regulatory kinase, induces BCAA oxidation primarily in skeletal muscle of healthy mice. While insulin acutely increases BCAA oxidation in cardiac and skeletal muscle, chronically insulin-resistant mice show blunted BCAA oxidation in adipose tissues and liver, shifting BCAA oxidation toward muscle. Together, this work provides a quantitative framework for understanding systemic BCAA oxidation in health and insulin resistance.
Collapse
Affiliation(s)
- Michael D Neinast
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Boulevard, Philadelphia, PA 19104, USA
| | - Cholsoon Jang
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Boulevard, Philadelphia, PA 19104, USA; Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Sheng Hui
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Danielle S Murashige
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Boulevard, Philadelphia, PA 19104, USA
| | - Qingwei Chu
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Boulevard, Philadelphia, PA 19104, USA
| | - Raphael J Morscher
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Xiaoxuan Li
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Le Zhan
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| | - Eileen White
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| | - Tracy G Anthony
- Department of Nutritional Sciences and the New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ 08901, USA
| | - Joshua D Rabinowitz
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Zoltan Arany
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Boulevard, Philadelphia, PA 19104, USA.
| |
Collapse
|
152
|
Mu W, Cheng XF, Liu Y, Lv QZ, Liu GL, Zhang JG, Li XY. Potential Nexus of Non-alcoholic Fatty Liver Disease and Type 2 Diabetes Mellitus: Insulin Resistance Between Hepatic and Peripheral Tissues. Front Pharmacol 2019; 9:1566. [PMID: 30692925 PMCID: PMC6339917 DOI: 10.3389/fphar.2018.01566] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/24/2018] [Indexed: 12/21/2022] Open
Abstract
The liver is the central metabolic organ and plays a pivotal role in regulating homeostasis of glucose and lipid metabolism. Aberrant liver metabolism promotes insulin resistance, which is reported to be a common characteristic of metabolic diseases such as non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM). There is a complex and bidirectional relationship between NAFLD and T2DM. NAFLD patients with hepatic insulin resistance generally share a high risk of impaired fasting glucose associated with early diabetes; most patients with T2DM experience non-alcoholic fatty liver (NAFL), non-alcoholic steatohepatitis (NASH), and other more severe liver complications such as cirrhosis and hepatocellular carcinoma (HCC). Additionally, hepatic insulin resistance, which is caused by diacylglycerol-mediated activation of protein kinase C epsilon (PKC𝜀), may be the critical pathological link between NAFLD and T2DM. Therefore, this review aims to illuminate current insights regarding the complex and strong association between NAFLD and T2DM and summarize novel and emerging targets for the treatment of hepatic insulin resistance based on established mechanistic knowledge.
Collapse
Affiliation(s)
- Wan Mu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue-Fang Cheng
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Liu
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qian-Zhou Lv
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gao-Lin Liu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ji-Gang Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Yu Li
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
153
|
Ren L, Zhou X, Huang X, Wang C, Li Y. The IRS/PI3K/Akt signaling pathway mediates olanzapine-induced hepatic insulin resistance in male rats. Life Sci 2018; 217:229-236. [PMID: 30550886 DOI: 10.1016/j.lfs.2018.12.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 11/19/2022]
Abstract
AIMS Chronic treatment with antipsychotics, especially most of atypical ones, leads to development of metabolic abnormalities. Olanzapine is an atypical antipsychotic widely used in the treatment of schizophrenia and bipolar disorder. The mechanisms underlying olanzapine-induced metabolic adverse effects in the liver, however, remain unclear. This study was designed to investigate olanzapine-induced insulin-desensitivity in the liver. MAIN METHODS Male rats were treated with olanzapine (5 mg/kg, by a gavage method, once daily for consecutive 8 weeks. Blood and liver variables were determined enzymatically or histologically. Gene/protein expression was analyzed by real-time PCR and Western blot. KEY FINDINGS Olanzapine treatment significantly increased fasting plasma insulin concentration, the index of the homeostasis model assessment of insulin resistance (HOMA-IR), and hepatic triglyceride and fatty droplet accumulation in rats. Hepatic gene/protein expression profile revealed that olanzapine activated mRNA and protein expression of sterol regulatory element-binding protein-1c, and mRNA levels of its downstream lipogenic enzymes, acetyl-CoA carboxylase-1, fatty acid synthase and stearoyl-CoA desaturase-1. More importantly, phosphorylated protein level of both Ser307 in insulin receptor substrate (IRS)-1 and Ser731 in IRS-2 was increased. Furthermore, phosphorylation of Tyr607 in phosphoinositide 3-kinase (PI3K) p85α, Ser473 in Akt and Ser2448 in mammalian target of rapamycin was also enhanced. SIGNIFICANCE Our results suggest that the IRS/PI3K/Akt signaling pathway mediates olanzapine-induced hepatic insulin resistance in male rats. Our findings may provide better understanding of the antipsychotic-induced metabolic adverse effects.
Collapse
Affiliation(s)
- Liying Ren
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xia Zhou
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoqian Huang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chunxia Wang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Yuhao Li
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Endocrinology and Metabolism Group, Sydney Institute of Health Sciences, Sydney Institute of Traditional Chinese Medicine, Sydney, NSW 2000, Australia.
| |
Collapse
|
154
|
Sharma R, Luong Q, Sharma VM, Harberson M, Harper B, Colborn A, Berryman DE, Jessen N, Jørgensen JOL, Kopchick JJ, Puri V, Lee KY. Growth hormone controls lipolysis by regulation of FSP27 expression. J Endocrinol 2018; 239:289-301. [PMID: 30400015 PMCID: PMC6226059 DOI: 10.1530/joe-18-0282] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 09/24/2018] [Indexed: 01/10/2023]
Abstract
Growth hormone (GH) has long been known to stimulate lipolysis and insulin resistance; however, the molecular mechanisms underlying these effects are unknown. In the present study, we demonstrate that GH acutely induces lipolysis in cultured adipocytes. This effect is secondary to the reduced expression of a negative regulator of lipolysis, fat-specific protein 27 (FSP27; aka Cidec) at both the mRNA and protein levels. These effects are mimicked in vivo as transgenic overexpression of GH leads to a reduction of FSP27 expression. Mechanistically, we show GH modulation of FSP27 expression is mediated through activation of both MEK/ERK- and STAT5-dependent intracellular signaling. These two molecular pathways interact to differentially manipulate peroxisome proliferator-activated receptor gamma activity (PPARγ) on the FSP27 promoter. Furthermore, overexpression of FSP27 is sufficient to fully suppress GH-induced lipolysis and insulin resistance in cultured adipocytes. Taken together, these data decipher a molecular mechanism by which GH acutely regulates lipolysis and insulin resistance in adipocytes.
Collapse
Affiliation(s)
- Rita Sharma
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Quyen Luong
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Vishva M. Sharma
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Mitchell Harberson
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Brian Harper
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Andrew Colborn
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Darlene E. Berryman
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Niels Jessen
- Research Laboratory for Biochemical Pathology, Aarhus University Hospital, Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Denmark
| | - Jens Otto Lunde Jørgensen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Medical Research Laboratory, Aarhus University, Aarhus, Denmark
| | - John J. Kopchick
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
- Edison Biotechnology Institute, Ohio University, Athens, OH
| | - Vishwajeet Puri
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| | - Kevin Y. Lee
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
- The Diabetes Institute, Ohio University, Athens, OH
| |
Collapse
|
155
|
El Ouaamari A, O-Sullivan I, Shirakawa J, Basile G, Zhang W, Roger S, Thomou T, Xu S, Qiang G, Liew CW, Kulkarni RN, Unterman TG. Forkhead box protein O1 (FoxO1) regulates hepatic serine protease inhibitor B1 (serpinB1) expression in a non-cell-autonomous fashion. J Biol Chem 2018; 294:1059-1069. [PMID: 30459233 DOI: 10.1074/jbc.ra118.006031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/15/2018] [Indexed: 12/13/2022] Open
Abstract
FoxO proteins are major targets of insulin action, and FoxO1 mediates the effects of insulin on hepatic glucose metabolism. We reported previously that serpinB1 is a liver-secreted factor (hepatokine) that promotes adaptive β-cell proliferation in response to insulin resistance in the liver-specific insulin receptor knockout (LIRKO) mouse. Here we report that FoxO1 plays a critical role in promoting serpinB1 expression in hepatic insulin resistance in a non-cell-autonomous manner. Mice lacking both the insulin receptor and FoxO1 (LIRFKO) exhibit reduced β-cell mass compared with LIRKO mice because of attenuation of β-cell proliferation. Although hepatic expression of serpinB1 mRNA and protein levels was increased in LIRKO mice, both the mRNA and protein levels returned to control levels in LIRFKO mice. Furthermore, liver-specific expression of constitutively active FoxO1 in transgenic mice induced an increase in hepatic serpinB1 mRNA and protein levels in refed mice. Conversely, serpinB1 mRNA and protein levels were reduced in mice lacking FoxO proteins in the liver. ChIP studies demonstrated that FoxO1 binds to three distinct sites located ∼9 kb upstream of the serpinb1 gene in primary mouse hepatocytes and that this binding is enhanced in hepatocytes from LIRKO mice. However, adenoviral expression of WT or constitutively active FoxO1 and insulin treatment are sufficient to regulate other FoxO1 target genes (IGFBP-1 and PEPCK) but not serpinB1 expression in mouse primary hepatocytes. These results indicate that liver FoxO1 promotes serpinB1 expression in hepatic insulin resistance and that non-cell-autonomous factors contribute to FoxO1-dependent effects on serpinB1 expression in the liver.
Collapse
Affiliation(s)
- Abdelfattah El Ouaamari
- From the Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Harvard Stem Cell Institute, Boston, Massachusetts 02215
| | - InSug O-Sullivan
- the Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois, Chicago, Illinois 60612.,the Medical Research Service, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| | - Jun Shirakawa
- From the Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Harvard Stem Cell Institute, Boston, Massachusetts 02215
| | - Giorgio Basile
- From the Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Harvard Stem Cell Institute, Boston, Massachusetts 02215
| | - Wenwei Zhang
- the Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois, Chicago, Illinois 60612.,the Medical Research Service, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| | - Sandra Roger
- From the Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Harvard Stem Cell Institute, Boston, Massachusetts 02215
| | - Thomas Thomou
- the Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02215, and
| | - Shanshan Xu
- the Department of Physiology and Biophysics, College of Medicine, University of Illinois, Chicago, Illinois 60612
| | - Guifen Qiang
- the Department of Physiology and Biophysics, College of Medicine, University of Illinois, Chicago, Illinois 60612
| | - Chong Wee Liew
- the Department of Physiology and Biophysics, College of Medicine, University of Illinois, Chicago, Illinois 60612
| | - Rohit N Kulkarni
- From the Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Harvard Stem Cell Institute, Boston, Massachusetts 02215,
| | - Terry G Unterman
- the Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois, Chicago, Illinois 60612, .,the Medical Research Service, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| |
Collapse
|
156
|
Santoleri D, Titchenell PM. Resolving the Paradox of Hepatic Insulin Resistance. Cell Mol Gastroenterol Hepatol 2018; 7:447-456. [PMID: 30739869 PMCID: PMC6369222 DOI: 10.1016/j.jcmgh.2018.10.016] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/30/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022]
Abstract
Insulin resistance is associated with numerous metabolic disorders, such as obesity and type II diabetes, that currently plague our society. Although insulin normally promotes anabolic metabolism in the liver by increasing glucose consumption and lipid synthesis, insulin-resistant individuals fail to inhibit hepatic glucose production and paradoxically have increased liver lipid synthesis, leading to hyperglycemia and hypertriglyceridemia. Here, we detail the intrahepatic and extrahepatic pathways mediating insulin's control of glucose and lipid metabolism. We propose that the interplay between both of these pathways controls insulin signaling and that mis-regulation between the 2 results in the paradoxic effects seen in the insulin-resistant liver instead of the commonly proposed deficiencies in particular branches of only the direct hepatic pathway.
Collapse
Affiliation(s)
- Dominic Santoleri
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Paul M. Titchenell
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania,Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania,Correspondence Address correspondence to: Paul M. Titchenell, PhD, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania 19104. fax: (215) 898-5408.
| |
Collapse
|
157
|
Wu Y, Pan Q, Yan H, Zhang K, Guo X, Xu Z, Yang W, Qi Y, Guo CA, Hornsby C, Zhang L, Zhou A, Li L, Chen Y, Zhang W, Sun Y, Zheng H, Wondisford F, He L, Guo S. Novel Mechanism of Foxo1 Phosphorylation in Glucagon Signaling in Control of Glucose Homeostasis. Diabetes 2018; 67:2167-2182. [PMID: 30201683 PMCID: PMC6198346 DOI: 10.2337/db18-0674] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/21/2018] [Indexed: 12/19/2022]
Abstract
Dysregulation of hepatic glucose production (HGP) serves as a major underlying mechanism for the pathogenesis of type 2 diabetes. The pancreatic hormone glucagon increases and insulin suppresses HGP, controlling blood glucose homeostasis. The forkhead transcription factor Foxo1 promotes HGP through increasing expression of genes encoding the rate-limiting enzymes responsible for gluconeogenesis. We previously established that insulin suppresses Foxo1 by Akt-mediated phosphorylation of Foxo1 at Ser256 in human hepatocytes. In this study, we found a novel Foxo1 regulatory mechanism by glucagon, which promotes Foxo1 nuclear translocation and stability via cAMP- and protein kinase A-dependent phosphorylation of Foxo1 at Ser276 Replacing Foxo1-S276 with alanine (A) or aspartate (D) to block or mimic phosphorylation, respectively, markedly regulates Foxo1 stability and nuclear localization in human hepatocytes. To establish in vivo function of Foxo1-Ser276 phosphorylation in glucose metabolism, we generated Foxo1-S273A and Foxo1-S273D knock-in (KI) mice. The KI mice displayed impaired blood glucose homeostasis, as well as the basal and glucagon-mediated HGP in hepatocytes. Thus, Foxo1-Ser276 is a new target site identified in the control of Foxo1 bioactivity and associated metabolic diseases.
Collapse
Affiliation(s)
- Yuxin Wu
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Quan Pan
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Hui Yan
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Kebin Zhang
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Xiaoqin Guo
- Department of Endocrinology, Third Military Medical University, Chongqing, China
- Division of Endocrinology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Zihui Xu
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Wanbao Yang
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Yajuan Qi
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Cathy A Guo
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Caitlyn Hornsby
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Lin Zhang
- Department of Chemistry, Cleveland State University, Cleveland, OH
| | - Aimin Zhou
- Department of Chemistry, Cleveland State University, Cleveland, OH
| | - Ling Li
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Yunmei Chen
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Weiping Zhang
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Yuxiang Sun
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Hongting Zheng
- Department of Endocrinology, Third Military Medical University, Chongqing, China
| | - Fred Wondisford
- Division of Endocrinology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Ling He
- Division of Endocrinology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Shaodong Guo
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| |
Collapse
|
158
|
Horst AK, Najjar SM, Wagener C, Tiegs G. CEACAM1 in Liver Injury, Metabolic and Immune Regulation. Int J Mol Sci 2018; 19:ijms19103110. [PMID: 30314283 PMCID: PMC6213298 DOI: 10.3390/ijms19103110] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/02/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is a transmembrane glycoprotein that is expressed on epithelial, endothelial and immune cells. CEACAM1 is a differentiation antigen involved in the maintenance of epithelial polarity that is induced during hepatocyte differentiation and liver regeneration. CEACAM1 regulates insulin sensitivity by promoting hepatic insulin clearance, and controls liver tolerance and mucosal immunity. Obese insulin-resistant humans with non-alcoholic fatty liver disease manifest loss of hepatic CEACAM1. In mice, deletion or functional inactivation of CEACAM1 impairs insulin clearance and compromises metabolic homeostasis which initiates the development of obesity and hepatic steatosis and fibrosis with other features of non-alcoholic steatohepatitis, and adipogenesis in white adipose depot. This is followed by inflammation and endothelial and cardiovascular dysfunctions. In obstructive and inflammatory liver diseases, soluble CEACAM1 is shed into human bile where it can serve as an indicator of liver disease. On immune cells, CEACAM1 acts as an immune checkpoint regulator, and deletion of Ceacam1 gene in mice causes exacerbation of inflammation and hyperactivation of myeloid cells and lymphocytes. Hence, hepatic CEACAM1 resides at the central hub of immune and metabolic homeostasis in both humans and mice. This review focuses on the regulatory role of CEACAM1 in liver and biliary tract architecture in health and disease, and on its metabolic role and function as an immune checkpoint regulator of hepatic inflammation.
Collapse
Affiliation(s)
- Andrea Kristina Horst
- Institute of Experimental Immunology and Hepatology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany.
| | - Sonia M Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Irvine Hall, 1 Ohio University, Athens, OH 45701-2979, USA.
- The Diabetes Institute, Heritage College of Osteopathic Medicine, Irvine Hall, 1 Ohio University, Athens, OH 45701-2979, USA.
| | - Christoph Wagener
- University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany.
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany.
| |
Collapse
|
159
|
Caracciolo V, Young J, Gonzales D, Ni Y, Flowers SJ, Summer R, Waldman SA, Kim JK, Jung DY, Noh HL, Kim T, Blackshear PJ, O'Connell D, Bauer RC, Kallen CB. Myeloid-specific deletion of Zfp36 protects against insulin resistance and fatty liver in diet-induced obese mice. Am J Physiol Endocrinol Metab 2018; 315:E676-E693. [PMID: 29509432 PMCID: PMC6230714 DOI: 10.1152/ajpendo.00224.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Obesity is associated with adipose tissue inflammation that contributes to insulin resistance. Zinc finger protein 36 (Zfp36) is an mRNA-binding protein that reduces inflammation by binding to cytokine transcripts and promoting their degradation. We hypothesized that myeloid-specific deficiency of Zfp36 would lead to increased adipose tissue inflammation and reduced insulin sensitivity in diet-induced obese mice. As expected, wild-type (Control) mice became obese and diabetic on a high-fat diet, and obese mice with myeloid-specific loss of Zfp36 [knockout (KO)] demonstrated increased adipose tissue and liver cytokine mRNA expression compared with Control mice. Unexpectedly, in glucose tolerance testing and hyperinsulinemic-euglycemic clamp studies, myeloid Zfp36 KO mice demonstrated improved insulin sensitivity compared with Control mice. Obese KO and Control mice had similar macrophage infiltration of the adipose depots and similar peripheral cytokine levels, but lean and obese KO mice demonstrated increased Kupffer cell (KC; the hepatic macrophage)-expressed Mac2 compared with lean Control mice. Insulin resistance in obese Control mice was associated with enhanced Zfp36 expression in KCs. Compared with Control mice, KO mice demonstrated increased hepatic mRNA expression of a multitude of classical (M1) inflammatory cytokines/chemokines, and this M1-inflammatory hepatic milieu was associated with enhanced nuclear localization of IKKβ and the p65 subunit of NF-κB. Our data confirm the important role of innate immune cells in regulating hepatic insulin sensitivity and lipid metabolism, challenge-prevailing models in which M1 inflammatory responses predict insulin resistance, and indicate that myeloid-expressed Zfp36 modulates the response to insulin in mice.
Collapse
Affiliation(s)
- Valentina Caracciolo
- Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Jeanette Young
- Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Donna Gonzales
- Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Yingchun Ni
- Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Stephen J Flowers
- Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Ross Summer
- Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Scott A Waldman
- Department of Pharmacology and Experimental Therapeutics, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Jason K Kim
- Program in Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Dae Young Jung
- Program in Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Hye Lim Noh
- Program in Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Taekyoon Kim
- Program in Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences , Research Triangle Park, North Carolina
| | - Danielle O'Connell
- Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Robert C Bauer
- Department of Medicine, Columbia University , New York, New York
| | - Caleb B Kallen
- Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| |
Collapse
|
160
|
Bergman RN, Piccinini F, Asare Bediako I, Kabir M, Kolka C, Polidori D, Ader M. Quantitative path to deep phenotyping: Possible importance of reduced hepatic insulin degradation to type 2 diabetes mellitus pathogenesis. J Diabetes 2018; 10:778-783. [PMID: 29961982 PMCID: PMC7219598 DOI: 10.1111/1753-0407.12794] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Diabetes is often thought of as one of two diseases: Type 1 diabetes (T1D), which is caused by immunological destruction of the beta-cells, and Type 2 diabetes (T2D), which is due to a combination of insulin resistance and relative failure of the beta-cells to compensate for the resistance. It is becoming clear, however, that even within these two definitions there may be considerable heterogeneity (1). There are several approaches to examine heterogeneity of T2D. Among these approaches are the use of biomarkers to categorize the disease, or the examination of variants in the genome. A third approach – the one we have been using in our laboratory – is to identify specific phenotypes which may contribute to failure to regulate the glucose level. We have identified a small group of such phenotypes which can be distinguished and measured using clinical protocols and/or mathematical modeling.
Collapse
Affiliation(s)
- Richard N Bergman
- Cedars-Sinai Medical Center, Sports Spectacular Diabetes and Obesity Wellness and Research Center, Los Angeles, California, USA
| | - Francesca Piccinini
- Cedars-Sinai Medical Center, Sports Spectacular Diabetes and Obesity Wellness and Research Center, Los Angeles, California, USA
| | - Isaac Asare Bediako
- Cedars-Sinai Medical Center, Sports Spectacular Diabetes and Obesity Wellness and Research Center, Los Angeles, California, USA
| | - Morvarid Kabir
- Cedars-Sinai Medical Center, Sports Spectacular Diabetes and Obesity Wellness and Research Center, Los Angeles, California, USA
| | - Cathryn Kolka
- Cedars-Sinai Medical Center, Sports Spectacular Diabetes and Obesity Wellness and Research Center, Los Angeles, California, USA
| | - David Polidori
- Janssen Research & Development, San Diego, California, USA
| | - Marilyn Ader
- Cedars-Sinai Medical Center, Sports Spectacular Diabetes and Obesity Wellness and Research Center, Los Angeles, California, USA
| |
Collapse
|
161
|
Petersen MC, Shulman GI. Mechanisms of Insulin Action and Insulin Resistance. Physiol Rev 2018; 98:2133-2223. [PMID: 30067154 PMCID: PMC6170977 DOI: 10.1152/physrev.00063.2017] [Citation(s) in RCA: 1697] [Impact Index Per Article: 242.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/22/2018] [Accepted: 03/24/2018] [Indexed: 12/15/2022] Open
Abstract
The 1921 discovery of insulin was a Big Bang from which a vast and expanding universe of research into insulin action and resistance has issued. In the intervening century, some discoveries have matured, coalescing into solid and fertile ground for clinical application; others remain incompletely investigated and scientifically controversial. Here, we attempt to synthesize this work to guide further mechanistic investigation and to inform the development of novel therapies for type 2 diabetes (T2D). The rational development of such therapies necessitates detailed knowledge of one of the key pathophysiological processes involved in T2D: insulin resistance. Understanding insulin resistance, in turn, requires knowledge of normal insulin action. In this review, both the physiology of insulin action and the pathophysiology of insulin resistance are described, focusing on three key insulin target tissues: skeletal muscle, liver, and white adipose tissue. We aim to develop an integrated physiological perspective, placing the intricate signaling effectors that carry out the cell-autonomous response to insulin in the context of the tissue-specific functions that generate the coordinated organismal response. First, in section II, the effectors and effects of direct, cell-autonomous insulin action in muscle, liver, and white adipose tissue are reviewed, beginning at the insulin receptor and working downstream. Section III considers the critical and underappreciated role of tissue crosstalk in whole body insulin action, especially the essential interaction between adipose lipolysis and hepatic gluconeogenesis. The pathophysiology of insulin resistance is then described in section IV. Special attention is given to which signaling pathways and functions become insulin resistant in the setting of chronic overnutrition, and an alternative explanation for the phenomenon of ‟selective hepatic insulin resistanceˮ is presented. Sections V, VI, and VII critically examine the evidence for and against several putative mediators of insulin resistance. Section V reviews work linking the bioactive lipids diacylglycerol, ceramide, and acylcarnitine to insulin resistance; section VI considers the impact of nutrient stresses in the endoplasmic reticulum and mitochondria on insulin resistance; and section VII discusses non-cell autonomous factors proposed to induce insulin resistance, including inflammatory mediators, branched-chain amino acids, adipokines, and hepatokines. Finally, in section VIII, we propose an integrated model of insulin resistance that links these mediators to final common pathways of metabolite-driven gluconeogenesis and ectopic lipid accumulation.
Collapse
Affiliation(s)
- Max C Petersen
- Departments of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine , New Haven, Connecticut
| | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine , New Haven, Connecticut
| |
Collapse
|
162
|
Rial SA, Ravaut G, Malaret TB, Bergeron KF, Mounier C. Hexanoic, Octanoic and Decanoic Acids Promote Basal and Insulin-Induced Phosphorylation of the Akt-mTOR Axis and a Balanced Lipid Metabolism in the HepG2 Hepatoma Cell Line. Molecules 2018; 23:molecules23092315. [PMID: 30208604 PMCID: PMC6225498 DOI: 10.3390/molecules23092315] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/06/2018] [Accepted: 09/08/2018] [Indexed: 12/29/2022] Open
Abstract
Metabolic illnesses such as non-alcoholic fatty liver disease (NAFLD) are in constant increase worldwide. Highly consumed long chain fatty acids (LCFA) are among the most obesogenic and steatogenic nutrients. Hepatic steatosis is associated with several complications such as insulin resistance. Growing evidence points to medium chain fatty acids (MCFA), more efficiently oxidized than LCFA, as a promising dietary alternative against NAFLD. However, reports on the hepatic effects of MCFA are sometimes conflicting. In this study we exposed HepG2 cells, a human hepatocellular model, to 0.25 mM of hexanoic (C6), or octanoic (C8), and decanoic (C10) acids separately or in a C8 + C10 equimolar mix reflecting commercially available MCFA-rich oils. We found that C6, a poorly studied MCFA, as well as C8 and C10 did not provoke the deleterious lipid anabolism runaway typically induced by LCFA palmitate. MCFA tended, instead, to promote a balanced metabolic profile and were generally non-cytotoxic. Accordingly, mitochondrial integrity was mostly preserved following MCFA treatment. However, treatments with C8 induced a mitochondrial membrane potential decrease, suggesting prolonged exposure to this lipid could be problematic. Finally, MCFA treatments maintained optimal insulin sensitivity and even fostered basal and insulin-dependent phosphorylation of the Akt-mTOR pathway. Overall, MCFA could constitute an effective nutritional tool to manage liver steatosis and hepatic insulin resistance.
Collapse
Affiliation(s)
- Sabri Ahmed Rial
- Molecular Metabolism of Lipids Laboratory, BioMed Research Center, Biological Sciences Department, University of Quebec in Montreal (UQAM), Montreal, QC H3C 3P8, Canada.
| | - Gaetan Ravaut
- Molecular Metabolism of Lipids Laboratory, BioMed Research Center, Biological Sciences Department, University of Quebec in Montreal (UQAM), Montreal, QC H3C 3P8, Canada.
| | - Tommy B Malaret
- Molecular Metabolism of Lipids Laboratory, BioMed Research Center, Biological Sciences Department, University of Quebec in Montreal (UQAM), Montreal, QC H3C 3P8, Canada.
| | - Karl-F Bergeron
- Molecular Metabolism of Lipids Laboratory, BioMed Research Center, Biological Sciences Department, University of Quebec in Montreal (UQAM), Montreal, QC H3C 3P8, Canada.
| | - Catherine Mounier
- Molecular Metabolism of Lipids Laboratory, BioMed Research Center, Biological Sciences Department, University of Quebec in Montreal (UQAM), Montreal, QC H3C 3P8, Canada.
| |
Collapse
|
163
|
Huang X, Liu G, Guo J, Su Z. The PI3K/AKT pathway in obesity and type 2 diabetes. Int J Biol Sci 2018; 14:1483-1496. [PMID: 30263000 PMCID: PMC6158718 DOI: 10.7150/ijbs.27173] [Citation(s) in RCA: 976] [Impact Index Per Article: 139.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/10/2018] [Indexed: 02/06/2023] Open
Abstract
Obesity and type 2 diabetes mellitus are complicated metabolic diseases that affect multiple organs and are characterized by hyperglycaemia. Currently, stable and effective treatments for obesity and type 2 diabetes mellitus are not available. Therefore, the mechanisms leading to obesity and diabetes and more effective ways to treat obesity and diabetes should be identified. Based on accumulated evidences, the PI3K/AKT signalling pathway is required for normal metabolism due to its characteristics, and its imbalance leads to the development of obesity and type 2 diabetes mellitus. This review focuses on the role of PI3K/AKT signalling in the skeletal muscle, adipose tissue, liver, brain and pancreas, and discusses how this signalling pathway affects the development of the aforementioned diseases. We also summarize evidences for recently identified therapeutic targets of the PI3K/AKT pathway as treatments for obesity and type 2 diabetes mellitus. PI3K/AKT pathway damaged in various tissues of the body leads to obesity and type 2 diabetes as the result of insulin resistance, and in turn, insulin resistance exacerbates the PI3K/AKT pathway, forming a vicious circle.
Collapse
Affiliation(s)
- Xingjun Huang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou (510006), China.,Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou (510006), China
| | - Guihua Liu
- Shenzhen Center for Disease Control and Prevention, 8 Longyuan Road, Nanshan District, Shenzhen (518055), China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou (510006), China
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou (510006), China.,Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou (510006), China
| |
Collapse
|
164
|
Asare-Bediako I, Paszkiewicz RL, Kim SP, Woolcott OO, Kolka CM, Burch MA, Kabir M, Bergman RN. Variability of Directly Measured First-Pass Hepatic Insulin Extraction and Its Association With Insulin Sensitivity and Plasma Insulin. Diabetes 2018; 67:1495-1503. [PMID: 29752425 PMCID: PMC6054441 DOI: 10.2337/db17-1520] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 05/03/2018] [Indexed: 01/20/2023]
Abstract
Although the β-cells secrete insulin, the liver, with its first-pass insulin extraction (FPE), regulates the amount of insulin allowed into circulation for action on target tissues. The metabolic clearance rate of insulin, of which FPE is the dominant component, is a major determinant of insulin sensitivity (SI). We studied the intricate relationship among FPE, SI, and fasting insulin. We used a direct method of measuring FPE, the paired portal/peripheral infusion protocol, where insulin is infused stepwise through either the portal vein or a peripheral vein in healthy young dogs (n = 12). FPE is calculated as the difference in clearance rates (slope of infusion rate vs. steady insulin plot) between the paired experiments. Significant correlations were found between FPE and clamp-assessed SI (rs = 0.74), FPE and fasting insulin (rs = -0.64), and SI and fasting insulin (rs = -0.67). We also found a wide variance in FPE (22.4-77.2%; mean ± SD 50.4 ± 19.1) that is reflected in the variability of plasma insulin (48.1 ± 30.9 pmol/L) and SI (9.4 ± 5.8 × 104 dL · kg-1 · min-1 · [pmol/L]-1). FPE could be the nexus of regulation of both plasma insulin and SI.
Collapse
Affiliation(s)
| | | | - Stella P Kim
- Cedars-Sinai Diabetes and Obesity Research Institute, Los Angeles, CA
| | - Orison O Woolcott
- Cedars-Sinai Diabetes and Obesity Research Institute, Los Angeles, CA
| | - Cathryn M Kolka
- Cedars-Sinai Diabetes and Obesity Research Institute, Los Angeles, CA
| | - Miguel A Burch
- Cedars-Sinai Medical Center, Department of Surgery, Los Angeles, CA
| | - Morvarid Kabir
- Cedars-Sinai Diabetes and Obesity Research Institute, Los Angeles, CA
| | - Richard N Bergman
- Cedars-Sinai Diabetes and Obesity Research Institute, Los Angeles, CA
| |
Collapse
|
165
|
Axen KV, Harper MA, Kuo YF, Axen K. Very low-carbohydrate, high-fat, weight reduction diet decreases hepatic gene response to glucose in obese rats. Nutr Metab (Lond) 2018; 15:54. [PMID: 31061673 PMCID: PMC6497366 DOI: 10.1186/s12986-018-0284-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 06/18/2018] [Indexed: 12/15/2022] Open
Abstract
Background Very low carbohydrate (VLC) diets are used to promote weight loss and improve insulin resistance (IR) in obesity. Since the high fat content of VLC diets may predispose to hepatic steatosis and hepatic insulin resistance, we investigated the effect of a VLC weight-reduction diet on measures of hepatic and whole body insulin resistance in obese rats. Methods In Phase 1, adult male Sprague-Dawley rats were made obese by ad libitum consumption of a high-fat (HF1, 60% of energy) diet; control rats ate a lower-fat (LF, 15%) diet for 10 weeks. In Phase 2, obese rats were fed energy-restricted amounts of a VLC (5%C, 65%F), LC (19%C, 55%F) or HC (55%C, 15%F) diet for 8 weeks while HF2 rats continued the HF diet ad libitum. In Phase 3, VLC rats were switched to the HC diet for 1 week. At the end of each phase, measurements of body composition and metabolic parameters were obtained. Hepatic insulin resistance was assessed by comparing expression of insulin-regulated genes following an oral glucose load,that increased plasma insulin levels, with the expression observed in the feed-deprived state. Results At the end of Phase 1, body weight, percent body fat, and hepatic lipid levels were greater in HF1 than LF rats (p < 0.05). At the end of Phase 2, percent body fat and intramuscular triglyceride decreased in LC and HC (p < 0.05), but not VLC rats, despite similar weight loss. VLC and HF2 rats had higher HOMA-IR and higher insulin at similar glucose levels following an ip glucose load than HC rats (p < 0.05). HC, but not VLC or HF2 rats, showed changes in Srebf1, Scd1, and Cpt1a expression (p < 0.05) in response to an oral glucose load. At the end of Phase 3, switching from the VLC to the HC diet mitigated differences in hepatic gene expression. Conclusion When compared with a high-carbohydrate, low-fat diet that produced similar weight loss, a commonly used VLC diet failed to improve whole body insulin resistance; it also reduced insulin’s effect on hepatic gene expression, which may reflect the development of hepatic insulin resistance.
Collapse
Affiliation(s)
- Kathleen V Axen
- Department of Health and Nutrition Sciences, Brooklyn College, City University of New York, New York, USA
| | - Marianna A Harper
- Department of Health and Nutrition Sciences, Brooklyn College, City University of New York, New York, USA
| | - Yu Fu Kuo
- Department of Health and Nutrition Sciences, Brooklyn College, City University of New York, New York, USA
| | - Kenneth Axen
- Department of Health and Nutrition Sciences, Brooklyn College, City University of New York, New York, USA
| |
Collapse
|
166
|
Fazakerley DJ, Krycer JR, Kearney AL, Hocking SL, James DE. Muscle and adipose tissue insulin resistance: malady without mechanism? J Lipid Res 2018; 60:1720-1732. [PMID: 30054342 DOI: 10.1194/jlr.r087510] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/25/2018] [Indexed: 12/14/2022] Open
Abstract
Insulin resistance is a major risk factor for numerous diseases, including type 2 diabetes and cardiovascular disease. These disorders have dramatically increased in incidence with modern life, suggesting that excess nutrients and obesity are major causes of "common" insulin resistance. Despite considerable effort, the mechanisms that contribute to common insulin resistance are not resolved. There is universal agreement that extracellular perturbations, such as nutrient excess, hyperinsulinemia, glucocorticoids, or inflammation, trigger intracellular stress in key metabolic target tissues, such as muscle and adipose tissue, and this impairs the ability of insulin to initiate its normal metabolic actions in these cells. Here, we present evidence that the impairment in insulin action is independent of proximal elements of the insulin signaling pathway and is likely specific to the glucoregulatory branch of insulin signaling. We propose that many intracellular stress pathways act in concert to increase mitochondrial reactive oxygen species to trigger insulin resistance. We speculate that this may be a physiological pathway to conserve glucose during specific states, such as fasting, and that, in the presence of chronic nutrient excess, this pathway ultimately leads to disease. This review highlights key points in this pathway that require further research effort.
Collapse
Affiliation(s)
- Daniel J Fazakerley
- School of Life and Environmental Sciences, Central Clinical School, University of Sydney, Camperdown, New South Wales, Australia
| | - James R Krycer
- School of Life and Environmental Sciences, Central Clinical School, University of Sydney, Camperdown, New South Wales, Australia
| | - Alison L Kearney
- School of Life and Environmental Sciences, Central Clinical School, University of Sydney, Camperdown, New South Wales, Australia
| | - Samantha L Hocking
- Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - David E James
- School of Life and Environmental Sciences, Central Clinical School, University of Sydney, Camperdown, New South Wales, Australia .,Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
167
|
Hepatic Dysfunction Caused by Consumption of a High-Fat Diet. Cell Rep 2018; 21:3317-3328. [PMID: 29241556 DOI: 10.1016/j.celrep.2017.11.059] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 11/11/2017] [Accepted: 11/16/2017] [Indexed: 12/16/2022] Open
Abstract
Obesity is a major human health crisis that promotes insulin resistance and, ultimately, type 2 diabetes. The molecular mechanisms that mediate this response occur across many highly complex biological regulatory levels that are incompletely understood. Here, we present a comprehensive molecular systems biology study of hepatic responses to high-fat feeding in mice. We interrogated diet-induced epigenomic, transcriptomic, proteomic, and metabolomic alterations using high-throughput omic methods and used a network modeling approach to integrate these diverse molecular signals. Our model indicated that disruption of hepatic architecture and enhanced hepatocyte apoptosis are among the numerous biological processes that contribute to early liver dysfunction and low-grade inflammation during the development of diet-induced metabolic syndrome. We validated these model findings with additional experiments on mouse liver sections. In total, we present an integrative systems biology study of diet-induced hepatic insulin resistance that uncovered molecular features promoting the development and maintenance of metabolic disease.
Collapse
|
168
|
Liao X, Song L, Zhang L, Wang H, Tong Q, Xu J, Yang G, Yang S, Zheng H. LAMP3 regulates hepatic lipid metabolism through activating PI3K/Akt pathway. Mol Cell Endocrinol 2018; 470:160-167. [PMID: 29056532 DOI: 10.1016/j.mce.2017.10.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/11/2017] [Accepted: 10/16/2017] [Indexed: 02/06/2023]
Abstract
Lysosome associated membrane protein 3 (LAMP3), a highly glycosylated protein, is one member of the LAMPs family. LAMPs family plays a critical role in the autolysosome fusion process. Autophagy was recently confirmed to regulate hepatic lipolysis. However, the physiological function of LAMP3 in lipid metabolism is not clear. In the current study, we discovered that the LAMP3 expression level was higher in the liver tissues of non-alcoholic fatty liver disease (NAFLD) patients and high-fat diet and ob/ob mice than in the matched control groups. LAMP3 expression was also obviously increased in hepatocellular carcinoma (HCC) cells treated with free fatty acids. Moreover, marked accumulation of intracellular lipid droplets and triglycerides (TG) was observed after LAMP3 overexpression in HCC cells. Further study showed that LAMP3 overexpression activated Akt and upregulated the expression of the lipogenic enzymes FASN and SCD-1 in HepG2 cells. Additionally, the increased TG content induced by LAMP3 overexpression was attenuated by treatment with a PI3K/Akt pathway inhibitor. Our findings demonstrated that LAMP3 is an important regulator of hepatic lipid metabolism, which provides a line of evidence for taking LAMP3 as a drug target in lipid metabolism disorder-associated diseases, such as NAFLD and obesity.
Collapse
Affiliation(s)
- Xiaoyu Liao
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Lingyu Song
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China; Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Linlin Zhang
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Hui Wang
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Qiang Tong
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jing Xu
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Gangyi Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| | - Hongting Zheng
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
169
|
Tao R, Wang C, Stöhr O, Qiu W, Hu Y, Miao J, Dong XC, Leng S, Stefater M, Stylopoulos N, Lin L, Copps KD, White MF. Inactivating hepatic follistatin alleviates hyperglycemia. Nat Med 2018; 24:1058-1069. [PMID: 29867232 PMCID: PMC6039237 DOI: 10.1038/s41591-018-0048-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 04/10/2018] [Indexed: 12/16/2022]
Abstract
Unsuppressed hepatic glucose production (HGP) contributes substantially to glucose intolerance and diabetes, which can be modeled by the genetic inactivation of hepatic insulin receptor substrate 1 (Irs1) and Irs2 (LDKO mice). We previously showed that glucose intolerance in LDKO mice is resolved by hepatic inactivation of the transcription factor FoxO1 (that is, LTKO mice)-even though the liver remains insensitive to insulin. Here, we report that insulin sensitivity in the white adipose tissue of LDKO mice is also impaired but is restored in LTKO mice in conjunction with normal suppression of HGP by insulin. To establish the mechanism by which white adipose tissue insulin signaling and HGP was regulated by hepatic FoxO1, we identified putative hepatokines-including excess follistatin (Fst)-that were dysregulated in LDKO mice but normalized in LTKO mice. Knockdown of hepatic Fst in the LDKO mouse liver restored glucose tolerance, white adipose tissue insulin signaling and the suppression of HGP by insulin; however, the expression of Fst in the liver of healthy LTKO mice had the opposite effect. Of potential clinical significance, knockdown of Fst also improved glucose tolerance in high-fat-fed obese mice, and the level of serum Fst was reduced in parallel with glycated hemoglobin in obese individuals with diabetes who underwent therapeutic gastric bypass surgery. We conclude that Fst is a pathological hepatokine that might be targeted for diabetes therapy during hepatic insulin resistance.
Collapse
Affiliation(s)
- Rongya Tao
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Caixia Wang
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oliver Stöhr
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wei Qiu
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yue Hu
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ji Miao
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - X Charlie Dong
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sining Leng
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Margaret Stefater
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicholas Stylopoulos
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lin Lin
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kyle D Copps
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Morris F White
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
170
|
Tian J, Wu J, Chen X, Guo T, Chen ZJ, Goldstein JL, Brown MS. BHLHE40, a third transcription factor required for insulin induction of SREBP-1c mRNA in rodent liver. eLife 2018; 7:36826. [PMID: 29952285 PMCID: PMC6023608 DOI: 10.7554/elife.36826] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/16/2018] [Indexed: 11/13/2022] Open
Abstract
In obesity, elevated insulin causes fatty liver by activating the gene encoding SREBP-1c, a transcription factor that enhances fatty acid synthesis. Two transcription factors, LXRα and C/EBPβ, are necessary but not sufficient for insulin induction of hepatic SREBP-1c mRNA. Here, we show that a third transcription factor, BHLHE40, is required. Immunoprecipitation revealed that BHLHE40 binds to C/EBPβ and LXRα in livers of rats that had fasted and then refed. Hepatic BHLHE40 mRNA rises rapidly when fasted rats are refed and when rat hepatocytes are incubated with insulin. Preventing this rise by gene knockout in mice or siRNAs in hepatocytes reduces the insulin-induced rise in SREBP-1c mRNA. Although BHLHE40 is necessary for insulin induction of SREBP-1c, it is not sufficient as demonstrated by failure of lentiviral BHLHE40 overexpression to increase hepatocyte SREBP-1c mRNA in the absence of insulin. Thus, an additional event is required for insulin to increase SREBP-1c mRNA.
Collapse
Affiliation(s)
- Jing Tian
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Jiaxi Wu
- Department of Molecular Biology and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Xiang Chen
- Department of Molecular Biology and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Tong Guo
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Zhijian J Chen
- Department of Molecular Biology and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Joseph L Goldstein
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Michael S Brown
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
171
|
Kubota H, Uda S, Matsuzaki F, Yamauchi Y, Kuroda S. In Vivo Decoding Mechanisms of the Temporal Patterns of Blood Insulin by the Insulin-AKT Pathway in the Liver. Cell Syst 2018; 7:118-128.e3. [PMID: 29960883 DOI: 10.1016/j.cels.2018.05.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/06/2018] [Accepted: 05/18/2018] [Indexed: 10/28/2022]
Abstract
Cells respond to various extracellular stimuli through a limited number of signaling pathways. One strategy to process such stimuli is to code the information into the temporal patterns of molecules. Although we showed that insulin selectively regulated molecules depending on its temporal patterns using Fao cells, the in vivo mechanism remains unknown. Here, we show how the insulin-AKT pathway processes the information encoded into the temporal patterns of blood insulin. We performed hyperinsulinemic-euglycemic clamp experiments and found that, in the liver, all temporal patterns of insulin are encoded into the insulin receptor, and downstream molecules selectively decode them through AKT. S6K selectively decodes the additional secretion information. G6Pase interprets the basal secretion information through FoxO1, while GSK3β decodes all secretion pattern information. Mathematical modeling revealed the mechanism via differences in network structures and from sensitivity and time constants. Given that almost all hormones exhibit distinct temporal patterns, temporal coding may be a general principle of system homeostasis by hormones.
Collapse
Affiliation(s)
- Hiroyuki Kubota
- Division of Integrated Omics, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan; PRESTO, Japan Science and Technology Agency, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan.
| | - Shinsuke Uda
- Division of Integrated Omics, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Fumiko Matsuzaki
- Division of Integrated Omics, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Yukiyo Yamauchi
- Division of Integrated Omics, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Shinya Kuroda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; CREST, Japan Science and Technology Corporation, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
172
|
White PJ, McGarrah RW, Grimsrud PA, Tso SC, Yang WH, Haldeman JM, Grenier-Larouche T, An J, Lapworth AL, Astapova I, Hannou SA, George T, Arlotto M, Olson LB, Lai M, Zhang GF, Ilkayeva O, Herman MA, Wynn RM, Chuang DT, Newgard CB. The BCKDH Kinase and Phosphatase Integrate BCAA and Lipid Metabolism via Regulation of ATP-Citrate Lyase. Cell Metab 2018; 27:1281-1293.e7. [PMID: 29779826 PMCID: PMC5990471 DOI: 10.1016/j.cmet.2018.04.015] [Citation(s) in RCA: 242] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 02/27/2018] [Accepted: 04/13/2018] [Indexed: 12/16/2022]
Abstract
Branched-chain amino acids (BCAA) are strongly associated with dysregulated glucose and lipid metabolism, but the underlying mechanisms are poorly understood. We report that inhibition of the kinase (BDK) or overexpression of the phosphatase (PPM1K) that regulates branched-chain ketoacid dehydrogenase (BCKDH), the committed step of BCAA catabolism, lowers circulating BCAA, reduces hepatic steatosis, and improves glucose tolerance in the absence of weight loss in Zucker fatty rats. Phosphoproteomics analysis identified ATP-citrate lyase (ACL) as an alternate substrate of BDK and PPM1K. Hepatic overexpression of BDK increased ACL phosphorylation and activated de novo lipogenesis. BDK and PPM1K transcript levels were increased and repressed, respectively, in response to fructose feeding or expression of the ChREBP-β transcription factor. These studies identify BDK and PPM1K as a ChREBP-regulated node that integrates BCAA and lipid metabolism. Moreover, manipulation of the BDK:PPM1K ratio relieves key metabolic disease phenotypes in a genetic model of severe obesity.
Collapse
Affiliation(s)
- Phillip J White
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA; Departments of Medicine and Pharmacology & Cancer Biology, Durham, NC 27701, USA
| | - Robert W McGarrah
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA; Departments of Medicine and Pharmacology & Cancer Biology, Durham, NC 27701, USA
| | - Paul A Grimsrud
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Shih-Chia Tso
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wen-Hsuan Yang
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Jonathan M Haldeman
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Thomas Grenier-Larouche
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Jie An
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Amanda L Lapworth
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Inna Astapova
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA; Departments of Medicine and Pharmacology & Cancer Biology, Durham, NC 27701, USA
| | - Sarah A Hannou
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Tabitha George
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Michelle Arlotto
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Lyra B Olson
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Michelle Lai
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA; Departments of Medicine and Pharmacology & Cancer Biology, Durham, NC 27701, USA
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Mark A Herman
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA; Departments of Medicine and Pharmacology & Cancer Biology, Durham, NC 27701, USA
| | - R Max Wynn
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David T Chuang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA; Departments of Medicine and Pharmacology & Cancer Biology, Durham, NC 27701, USA.
| |
Collapse
|
173
|
Varghese J, James J, Vaulont S, Mckie A, Jacob M. Increased intracellular iron in mouse primary hepatocytes in vitro causes activation of the Akt pathway but decreases its response to insulin. Biochim Biophys Acta Gen Subj 2018; 1862:1870-1882. [PMID: 29859963 DOI: 10.1016/j.bbagen.2018.05.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/17/2018] [Accepted: 05/28/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND An iron-overloaded state has been reported to be associated with insulin resistance. On the other hand, conditions such as classical hemochromatosis (where iron overload occurs primarily in the liver) have been reported to be associated with increased insulin sensitivity. The reasons for these contradictory findings are unclear. In this context, the effects of increased intracellular iron per se on insulin signaling in hepatocytes are not known. METHODS Mouse primary hepatocytes were loaded with iron in vitro by incubation with ferric ammonium citrate (FAC). Intracellular events related to insulin signaling, as well as changes in gene expression and hepatocyte glucose production (HGP), were studied in the presence and absence of insulin and/or forskolin (a glucagon mimetic). RESULTS In vitro iron-loading of hepatocytes resulted in phosphorylation-mediated activation of Akt and AMP-activated protein kinase. This was associated with decreased basal and forskolin-stimulated HGP. Iron attenuated forskolin-mediated induction of the key gluconeogenic enzyme, glucose-6-phosphatase. It also attenuated activation of the Akt pathway in response to insulin, which was associated with decreased protein levels of insulin receptor substrates 1 and 2, constituting insulin resistance. CONCLUSIONS Increased intracellular iron has dual effects on insulin sensitivity in hepatocytes. It increased basal activation of the Akt pathway, but decreased activation of this pathway in response to insulin. GENERAL SIGNIFICANCE These findings may help explain why both insulin resistance and increased sensitivity have been observed in iron-overloaded states. They are of relevance to a variety of disease conditions characterized by hepatic iron overload and increased risk of diabetes.
Collapse
Affiliation(s)
- Joe Varghese
- Department of Biochemistry, Christian Medical College, Vellore 632002, India(1).
| | - Jithu James
- Department of Biochemistry, Christian Medical College, Vellore 632002, India(1)
| | | | - Andrew Mckie
- Diabetes and Nutritional Sciences Division, School of Medicine, King's College, London, UK
| | - Molly Jacob
- Department of Biochemistry, Christian Medical College, Vellore 632002, India(1)
| |
Collapse
|
174
|
Ginsenoside Rb2 promotes glucose metabolism and attenuates fat accumulation via AKT-dependent mechanisms. Biomed Pharmacother 2018; 100:93-100. [DOI: 10.1016/j.biopha.2018.01.111] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/21/2018] [Accepted: 01/24/2018] [Indexed: 12/29/2022] Open
|
175
|
Miller RA, Shi Y, Lu W, Pirman DA, Jatkar A, Blatnik M, Wu H, Cárdenas C, Wan M, Foskett JK, Park JO, Zhang Y, Holland WL, Rabinowitz JD, Birnbaum MJ. Targeting hepatic glutaminase activity to ameliorate hyperglycemia. Nat Med 2018; 24:518-524. [PMID: 29578539 PMCID: PMC6089616 DOI: 10.1038/nm.4514] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 02/08/2018] [Indexed: 02/07/2023]
Affiliation(s)
- Russell A Miller
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Pfizer Internal Medicine Research Units, Cambridge, Massachusetts, USA
| | - Yuji Shi
- Pfizer Internal Medicine Research Units, Cambridge, Massachusetts, USA
| | - Wenyun Lu
- Chemistry and Integrative Genomics, Princeton University, Princeton, New Jersey, USA
| | - David A Pirman
- Pfizer Worldwide Research and Development, Groton, Connecticut, USA
| | - Aditi Jatkar
- Pfizer Internal Medicine Research Units, Cambridge, Massachusetts, USA
| | - Matthew Blatnik
- Pfizer Worldwide Research and Development, Groton, Connecticut, USA
| | - Hong Wu
- Pfizer Worldwide Research and Development, Groton, Connecticut, USA
| | - César Cárdenas
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,Buck Institute for Research on Aging, Novato, California, USA.,Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, California, USA
| | - Min Wan
- Pfizer Internal Medicine Research Units, Cambridge, Massachusetts, USA
| | - J Kevin Foskett
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Junyoung O Park
- Chemistry and Integrative Genomics, Princeton University, Princeton, New Jersey, USA.,Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, USA
| | - Yiyi Zhang
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - William L Holland
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joshua D Rabinowitz
- Chemistry and Integrative Genomics, Princeton University, Princeton, New Jersey, USA
| | - Morris J Birnbaum
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Pfizer Internal Medicine Research Units, Cambridge, Massachusetts, USA.,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
176
|
Key CCC, Liu M, Kurtz CL, Chung S, Boudyguina E, Dinh TA, Bashore A, Phelan PE, Freedman BI, Osborne TF, Zhu X, Ma L, Sethupathy P, Biddinger SB, Parks JS. Hepatocyte ABCA1 Deletion Impairs Liver Insulin Signaling and Lipogenesis. Cell Rep 2018; 19:2116-2129. [PMID: 28591582 DOI: 10.1016/j.celrep.2017.05.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 03/07/2017] [Accepted: 05/09/2017] [Indexed: 10/19/2022] Open
Abstract
Plasma membrane (PM) free cholesterol (FC) is emerging as an important modulator of signal transduction. Here, we show that hepatocyte-specific knockout (HSKO) of the cellular FC exporter, ATP-binding cassette transporter A1 (ABCA1), leads to decreased PM FC content and defective trafficking of lysosomal FC to the PM. Compared with controls, chow-fed HSKO mice had reduced hepatic (1) insulin-stimulated Akt phosphorylation, (2) activation of the lipogenic transcription factor Sterol Regulatory Element Binding Protein (SREBP)-1c, and (3) lipogenic gene expression. Consequently, Western-type diet-fed HSKO mice were protected from steatosis. Surprisingly, HSKO mice had intact glucose metabolism; they showed normal gluconeogenic gene suppression in response to re-feeding and normal glucose and insulin tolerance. We conclude that: (1) ABCA1 maintains optimal hepatocyte PM FC, through intracellular FC trafficking, for efficient insulin signaling; and (2) hepatocyte ABCA1 deletion produces a form of selective insulin resistance so that lipogenesis is suppressed but glucose metabolism remains normal.
Collapse
Affiliation(s)
- Chia-Chi C Key
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Mingxia Liu
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - C Lisa Kurtz
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Soonkyu Chung
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE 68588, USA
| | - Elena Boudyguina
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Timothy A Dinh
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alexander Bashore
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Peter E Phelan
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Barry I Freedman
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Timothy F Osborne
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Xuewei Zhu
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Lijun Ma
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Praveen Sethupathy
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sudha B Biddinger
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02062, USA
| | - John S Parks
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
177
|
Ling AV, Gearing ME, Semova I, Shin DJ, Clements R, Lai ZW, Biddinger SB. FoxO1 Is Required for Most of the Metabolic and Hormonal Perturbations Produced by Hepatic Insulin Receptor Deletion in Male Mice. Endocrinology 2018; 159:1253-1263. [PMID: 29300910 PMCID: PMC5802805 DOI: 10.1210/en.2017-00870] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 12/21/2017] [Indexed: 12/16/2022]
Abstract
Insulin coordinates the complex response to feeding, affecting numerous metabolic and hormonal pathways. Forkhead box protein O1 (FoxO1) is one of several signaling molecules downstream of insulin; FoxO1 drives gluconeogenesis and is suppressed by insulin. To determine the role of FoxO1 in mediating other actions of insulin, we studied mice with hepatic deletion of the insulin receptor, FoxO1, or both. We found that mice with deletion of the insulin receptor alone showed not only hyperglycemia but also a 70% decrease in plasma insulin-like growth factor 1 and delayed growth during the first 2 months of life, a 24-fold increase in the soluble leptin receptor and a 19-fold increase in plasma leptin levels. Deletion of the insulin receptor also produced derangements in fatty acid metabolism, with a decrease in the expression of the lipogenic enzymes, hepatic diglycerides, and plasma triglycerides; in parallel, it increased expression of the fatty acid oxidation enzymes. Mice with deletion of both insulin receptor and FoxO1 showed a much more modest phenotype, with normal or near-normal glucose levels, growth, leptin levels, hepatic diglycerides, and fatty acid oxidation gene expression; however, lipogenic gene expression remained low. Taken together, these data reveal the pervasive role of FoxO1 in mediating the effects of insulin on not only glucose metabolism but also other hormonal signaling pathways and even some aspects of lipid metabolism.
Collapse
Affiliation(s)
- Alisha V. Ling
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Mary E. Gearing
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Ivana Semova
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Dong-Ju Shin
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Rebecca Clements
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Zon W. Lai
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115
| | - Sudha B. Biddinger
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
178
|
Zhang Y, Meng F, Sun X, Sun X, Hu M, Cui P, Vestin E, Li X, Li W, Wu XK, Jansson JO, Shao LR, Billig H. Hyperandrogenism and insulin resistance contribute to hepatic steatosis and inflammation in female rat liver. Oncotarget 2018; 9:18180-18197. [PMID: 29719598 PMCID: PMC5915065 DOI: 10.18632/oncotarget.24477] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/25/2018] [Indexed: 02/06/2023] Open
Abstract
Women with polycystic ovary syndrome (PCOS) are at high risk for nonalcoholic fatty liver disease (NAFLD). While insulin resistance is a common trait for both PCOS and NAFLD, hyperandrogenism is also considered to be a key factor contributing to PCOS, and the molecular mechanisms behind the interactions between insulin resistance and hyperandrogenism in the female liver remain largely unexplored. Using chronic treatment with insulin and/or human chorionic gonadotropin (hCG), we showed that all female rats with different treatments induced imbalance between de novo lipogenesis and mitochondrial β-oxidation via the Pparα/β–Srebp1/2–Acc1 axis, resulting in varying degrees of hepatic steatosis. Given the fact that hepatic lipid metabolism and inflammation are tightly linked processes, we found that hCG-induced hyperandrogenic rats had strongly aggravated hepatic inflammation. Further mechanistic investigations revealed that dysregulation of the IRS–PI3K–Akt signaling axis that integrated aberrant inflammatory, apoptotic and autophagic responses in the liver was strongly associated with hyperandrogenism itself or combined with insulin resistance. Additionally, we found that hCG-treated and insulin+hCG-induced rats developed visceral adipose tissue inflammation characterized by the presence of “crown like” structure and increased inflammatory gene expression. Because a more pronounced hepatic steatosis, inflammatory responses, and hepatocyte cell damage were observed in insulin+hCG-induced PCOS-like rats, our finding suggest that NAFLD seen in PCOS patients is dependent of hyperandrogenism and insulin resistance.
Collapse
Affiliation(s)
- Yuehui Zhang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China.,Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Fanci Meng
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - Xiaoyan Sun
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - Xue Sun
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - Min Hu
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Peng Cui
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden.,Department of Integrative Medicine and Neurobiology, State Key Lab of Medical Neurobiology, Shanghai Medical College and Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institute of Brain Science, Fudan University, 200032 Shanghai, China.,Institute of Integrative Medicine of Fudan University, 200032 Shanghai, China
| | - Edvin Vestin
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Xin Li
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden.,Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 200011 Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, 200011 Shanghai, China
| | - Wei Li
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - Xiao-Ke Wu
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - John-Olov Jansson
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Linus R Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Håkan Billig
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| |
Collapse
|
179
|
Insulin selectively reduces mitochondrial uncoupling in brown adipose tissue in mice. Biochem J 2018; 475:561-569. [PMID: 29170160 DOI: 10.1042/bcj20170736] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/20/2017] [Accepted: 11/22/2017] [Indexed: 12/17/2022]
Abstract
The purpose of the present study was to determine the effects of prolonged hyperinsulinemia on mitochondrial respiration and uncoupling in distinct adipose tissue depots. Sixteen-week-old male mice were injected daily with placebo or insulin to induce an artificial hyperinsulinemia for 28 days. Following the treatment period, mitochondrial respiration and degree of uncoupling were determined in permeabilized perirenal, inguinal, and interscapular adipose tissue. White adipose tissue (WAT) mitochondria (inguinal and perirenal) respire at substantially lower rates compared with brown adipose tissue (BAT). Insulin treatment resulted in a significant reduction in mitochondrial respiration in inguinal WAT (iWAT) and interscapular BAT (iBAT), but not in perirenal WAT (pWAT). Furthermore, these changes were accompanied by an insulin-induced reduction in UCP-1 (uncoupling protein 1) and PGC-1α in iWAT and iBAT only, but not in pWAT or skeletal muscle. Compared with adipose tissue mitochondria in placebo conditions, adipose tissue from hyperinsulinemic mice manifested a site-specific reduction in mitochondrial respiration probably as a result of reduced uncoupling. These results may help explain weight gain so commonly seen with insulin treatment in type 2 diabetes mellitus.
Collapse
|
180
|
Fazakerley DJ, Chaudhuri R, Yang P, Maghzal GJ, Thomas KC, Krycer JR, Humphrey SJ, Parker BL, Fisher-Wellman KH, Meoli CC, Hoffman NJ, Diskin C, Burchfield JG, Cowley MJ, Kaplan W, Modrusan Z, Kolumam G, Yang JY, Chen DL, Samocha-Bonet D, Greenfield JR, Hoehn KL, Stocker R, James DE. Mitochondrial CoQ deficiency is a common driver of mitochondrial oxidants and insulin resistance. eLife 2018; 7:32111. [PMID: 29402381 PMCID: PMC5800848 DOI: 10.7554/elife.32111] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/02/2018] [Indexed: 12/11/2022] Open
Abstract
Insulin resistance in muscle, adipocytes and liver is a gateway to a number of metabolic diseases. Here, we show a selective deficiency in mitochondrial coenzyme Q (CoQ) in insulin-resistant adipose and muscle tissue. This defect was observed in a range of in vitro insulin resistance models and adipose tissue from insulin-resistant humans and was concomitant with lower expression of mevalonate/CoQ biosynthesis pathway proteins in most models. Pharmacologic or genetic manipulations that decreased mitochondrial CoQ triggered mitochondrial oxidants and insulin resistance while CoQ supplementation in either insulin-resistant cell models or mice restored normal insulin sensitivity. Specifically, lowering of mitochondrial CoQ caused insulin resistance in adipocytes as a result of increased superoxide/hydrogen peroxide production via complex II. These data suggest that mitochondrial CoQ is a proximal driver of mitochondrial oxidants and insulin resistance, and that mechanisms that restore mitochondrial CoQ may be effective therapeutic targets for treating insulin resistance. After we eat, our blood sugar levels increase. To counteract this, the pancreas releases a hormone called insulin. Part of insulin’s effect is to promote the uptake of sugar from the blood into muscle and fat tissue for storage. Under certain conditions, such as obesity, this process can become defective, leading to a condition known as insulin resistance. This condition makes a number of human diseases more likely to develop, including type 2 diabetes. Working out how insulin resistance develops could therefore unveil new treatment strategies for these diseases. Mitochondria – structures that produce most of a cell’s energy supply – appear to play a role in the development of insulin resistance. Mitochondria convert nutrients such as fats and sugars into molecules called ATP that fuel the many processes required for life. However, ATP production can also generate potentially harmful intermediates often referred to as ‘reactive oxygen species’ or ‘oxidants’. Previous studies have suggested that an increase in the amount of oxidants produced in mitochondria can cause insulin resistance. Fazakerley et al. therefore set out to identify the reason for increased oxidants in mitochondria, and did so by analysing the levels of proteins and oxidants found in cells grown in the laboratory, and mouse and human tissue samples. This led them to find that concentrations of a molecule called coenzyme Q (CoQ), an essential component of mitochondria that helps to produce ATP, were lower in mitochondria from insulin-resistant fat and muscle tissue. Further experiments suggested a link between the lower levels of CoQ and the higher levels of oxidants in the mitochondria. Replenishing the mitochondria of the lab-grown cells and insulin-resistant mice with CoQ restored ‘normal’ oxidant levels and prevented the development of insulin resistance. Strategies that aim to increase mitochondria CoQ levels may therefore prevent or reverse insulin resistance. Although CoQ supplements are readily available, swallowing CoQ does not efficiently deliver CoQ to mitochondria in humans, so alternative treatment methods must be found. It is also of interest that statins, common drugs taken by millions of people around the world to lower cholesterol, also lower CoQ and have been reported to increase the risk of developing type 2 diabetes. Further research is therefore needed to investigate whether CoQ might provide the link between statins and type 2 diabetes.
Collapse
Affiliation(s)
- Daniel J Fazakerley
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Rima Chaudhuri
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Pengyi Yang
- School of Mathematics and Statistics, University of Sydney, Camperdown, Australia
| | - Ghassan J Maghzal
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Kristen C Thomas
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - James R Krycer
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Sean J Humphrey
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Benjamin L Parker
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Kelsey H Fisher-Wellman
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, United States
| | - Christopher C Meoli
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Nolan J Hoffman
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Ciana Diskin
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - James G Burchfield
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Mark J Cowley
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Warren Kaplan
- Peter Wills Bioinformatics Centre, Garvan Institute of Medical Research, Darlinghurst, Australia
| | | | | | - Jean Yh Yang
- School of Mathematics and Statistics, University of Sydney, Camperdown, Australia
| | - Daniel L Chen
- Garvan Institute of Medical Research, Darlinghurst, Australia
| | | | | | - Kyle L Hoehn
- School of Biotechnology and Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Roland Stocker
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia.,Charles Perkins Centre, Sydney Medical School, University of Sydney, Camperdown NSW, Australia
| |
Collapse
|
181
|
Tubbs E, Axelsson AS, Vial G, Wollheim CB, Rieusset J, Rosengren AH. Sulforaphane improves disrupted ER-mitochondria interactions and suppresses exaggerated hepatic glucose production. Mol Cell Endocrinol 2018; 461:205-214. [PMID: 28923347 DOI: 10.1016/j.mce.2017.09.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/10/2017] [Accepted: 09/13/2017] [Indexed: 02/07/2023]
Abstract
AIMS Exaggerated hepatic glucose production is one of the hallmarks of type 2 diabetes. Sulforaphane (SFN) has been suggested as a new potential anti-diabetic compound. However, the effects of SFN in hepatocytes are yet unclear. Accumulating evidence points to the close structural contacts between the ER and mitochondria, known as mitochondria-associated ER membranes (MAMs), as important hubs for hepatic metabolism. We wanted to investigate whether SFN could affect hepatic glucose production and MAMs. MATERIALS AND METHODS We used proximity ligation assays, analysis of ER stress markers and glucose production assays in hepatoma cell lines, primary mouse hepatocytes and diabetic animal models. RESULTS SFN counteracted the increase of glucose production in palmitate-treated mouse hepatocytes. SFN also counteracted palmitate-induced MAM disruptions. Moreover, SFN decreased the ER stress markers CHOP and Grp78. In ob/ob mice, SFN improved glucose tolerance and reduced exaggerated glucose production. In livers of these mice, SFN increased MAM protein content, restored impaired VDAC1-IP3R1 interactions and reduced ER stress markers. In mice on HFHSD, SFN improved glucose tolerance, MAM protein content and ER-mitochondria interactions to a similar extent to that of metformin. CONCLUSIONS The present findings show that MAMs are severely reduced in animal models of glucose intolerance, which reinforces the role of MAMs as a hub for insulin signaling in the liver. We also show that SFN restores MAMs and improves glucose tolerance by a similar magnitude to that of metformin. These data highlight SFN as a new potential anti-diabetic compound.
Collapse
Affiliation(s)
- Emily Tubbs
- Lund University Diabetes Centre, Jan Waldenströms Gata 35, SE-20502 Malmö, Sweden
| | - Annika S Axelsson
- Lund University Diabetes Centre, Jan Waldenströms Gata 35, SE-20502 Malmö, Sweden
| | - Guillaume Vial
- INSERM UMR-1060 CarMeN Laboratory, Lyon 1 University, INRA U1235, INSA of Lyon, Charles Merieux Lyon-Sud Medical Universities, Lyon, France
| | - Claes B Wollheim
- Lund University Diabetes Centre, Jan Waldenströms Gata 35, SE-20502 Malmö, Sweden; Department of Cell Physiology and Metabolism, University Medical Center, 1211 Geneva, Switzerland
| | - Jennifer Rieusset
- INSERM UMR-1060 CarMeN Laboratory, Lyon 1 University, INRA U1235, INSA of Lyon, Charles Merieux Lyon-Sud Medical Universities, Lyon, France
| | - Anders H Rosengren
- Lund University Diabetes Centre, Jan Waldenströms Gata 35, SE-20502 Malmö, Sweden; Department of Neuroscience and Physiology, University of Gothenburg, Medicinaregatan 11, SE-41390 Göteborg, Sweden.
| |
Collapse
|
182
|
Corbit KC, Camporez JPG, Edmunds LR, Tran JL, Vera NB, Erion DM, Deo RC, Perry RJ, Shulman GI, Jurczak MJ, Weiss EJ. Adipocyte JAK2 Regulates Hepatic Insulin Sensitivity Independently of Body Composition, Liver Lipid Content, and Hepatic Insulin Signaling. Diabetes 2018; 67:208-221. [PMID: 29203511 PMCID: PMC5780061 DOI: 10.2337/db17-0524] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 11/15/2017] [Indexed: 01/07/2023]
Abstract
Disruption of hepatocyte growth hormone (GH) signaling through disruption of Jak2 (JAK2L) leads to fatty liver. Previously, we demonstrated that development of fatty liver depends on adipocyte GH signaling. We sought to determine the individual roles of hepatocyte and adipocyte Jak2 on whole-body and tissue insulin sensitivity and liver metabolism. On chow, JAK2L mice had hepatic steatosis and severe whole-body and hepatic insulin resistance. However, concomitant deletion of Jak2 in hepatocytes and adipocytes (JAK2LA) completely normalized insulin sensitivity while reducing liver lipid content. On high-fat diet, JAK2L mice had hepatic steatosis and insulin resistance despite protection from diet-induced obesity. JAK2LA mice had higher liver lipid content and no protection from obesity but retained exquisite hepatic insulin sensitivity. AKT activity was selectively attenuated in JAK2L adipose tissue, whereas hepatic insulin signaling remained intact despite profound hepatic insulin resistance. Therefore, JAK2 in adipose tissue is epistatic to liver with regard to insulin sensitivity and responsiveness, despite fatty liver and obesity. However, hepatocyte autonomous JAK2 signaling regulates liver lipid deposition under conditions of excess dietary fat. This work demonstrates how various tissues integrate JAK2 signals to regulate insulin/glucose and lipid metabolism.
Collapse
Affiliation(s)
- Kevin C Corbit
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | | | - Lia R Edmunds
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Jennifer L Tran
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Nicholas B Vera
- Cardiovascular and Metabolic Diseases, Pfizer, Cambridge, MA
| | - Derek M Erion
- Cardiovascular and Metabolic Diseases, Pfizer, Cambridge, MA
| | - Rahul C Deo
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Rachel J Perry
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
| | - Michael J Jurczak
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Ethan J Weiss
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
183
|
Unterman TG. Regulation of Hepatic Glucose Metabolism by FoxO Proteins, an Integrated Approach. Curr Top Dev Biol 2018; 127:119-147. [DOI: 10.1016/bs.ctdb.2017.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
184
|
Guri Y, Colombi M, Dazert E, Hindupur SK, Roszik J, Moes S, Jenoe P, Heim MH, Riezman I, Riezman H, Hall MN. mTORC2 Promotes Tumorigenesis via Lipid Synthesis. Cancer Cell 2017; 32:807-823.e12. [PMID: 29232555 DOI: 10.1016/j.ccell.2017.11.011] [Citation(s) in RCA: 289] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 08/06/2017] [Accepted: 11/14/2017] [Indexed: 12/22/2022]
Abstract
Dysregulated mammalian target of rapamycin (mTOR) promotes cancer, but underlying mechanisms are poorly understood. We describe an mTOR-driven mouse model that displays hepatosteatosis progressing to hepatocellular carcinoma (HCC). Longitudinal proteomic, lipidomics, and metabolomic analyses revealed that hepatic mTORC2 promotes de novo fatty acid and lipid synthesis, leading to steatosis and tumor development. In particular, mTORC2 stimulated sphingolipid (glucosylceramide) and glycerophospholipid (cardiolipin) synthesis. Inhibition of fatty acid or sphingolipid synthesis prevented tumor development, indicating a causal effect in tumorigenesis. Increased levels of cardiolipin were associated with tubular mitochondria and enhanced oxidative phosphorylation. Furthermore, increased lipogenesis correlated with elevated mTORC2 activity and HCC in human patients. Thus, mTORC2 promotes cancer via formation of lipids essential for growth and energy production.
Collapse
Affiliation(s)
- Yakir Guri
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Marco Colombi
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Eva Dazert
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | | | - Jason Roszik
- Departments of Melanoma Medical Oncology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Suzette Moes
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Paul Jenoe
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Markus H Heim
- Department of Biomedicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Isabelle Riezman
- NCCR Chemical Biology, Department of Biochemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Howard Riezman
- NCCR Chemical Biology, Department of Biochemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Michael N Hall
- Biozentrum, University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
185
|
Hu Y, Semova I, Sun X, Kang H, Chahar S, Hollenberg AN, Masson D, Hirschey MD, Miao J, Biddinger SB. Fructose and glucose can regulate mammalian target of rapamycin complex 1 and lipogenic gene expression via distinct pathways. J Biol Chem 2017; 293:2006-2014. [PMID: 29222328 DOI: 10.1074/jbc.m117.782557] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 11/20/2017] [Indexed: 12/19/2022] Open
Abstract
Although calorically equivalent to glucose, fructose appears to be more lipogenic, promoting dyslipidemia, fatty liver disease, cardiovascular disease, and diabetes. To better understand how fructose induces lipogenesis, we compared the effects of fructose and glucose on mammalian target of rapamycin complex 1 (mTORC1), which appeared to have both positive and negative effects on lipogenic gene expression. We found that fructose acutely and transiently suppressed mTORC1 signaling in vitro and in vivo The constitutive activation of mTORC1 reduced hepatic lipogenic gene expression and produced hypotriglyceridemia after 1 week of fructose feeding. In contrast, glucose did not suppress mTORC1, and the constitutive activation of mTORC1 failed to suppress plasma triglycerides after 1 week of glucose feeding. Thus, these data reveal fundamental differences in the signaling pathways used by fructose and glucose to regulate lipid metabolism.
Collapse
Affiliation(s)
- Yue Hu
- From the Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Ivana Semova
- From the Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Xiaowei Sun
- From the Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Hong Kang
- From the Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Satyapal Chahar
- From the Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Anthony N Hollenberg
- Division of Endocrinology, Metabolism and Diabetes, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - David Masson
- INSERM Lipid Nutrition Cancer UMR 1231, 21000 Dijon, France, and
| | - Matthew D Hirschey
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina 27710
| | - Ji Miao
- From the Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115,
| | - Sudha B Biddinger
- From the Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115,
| |
Collapse
|
186
|
Ramakrishnan SK, Shah YM. A central role for hypoxia-inducible factor (HIF)-2α in hepatic glucose homeostasis. ACTA ACUST UNITED AC 2017; 4:207-216. [PMID: 29276790 PMCID: PMC5734117 DOI: 10.3233/nha-170022] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hepatic glucose production is regulated by hormonal and dietary factors. At fasting, 80% of glucose released into the circulation is derived from the liver, among which gluconeogenesis accounts for 55% and the rest by glycogenolysis. Studies suggest a complex mechanism involved in the regulation of hepatic glucose metabolism during fasting and post-absorptive phase. Oxygen plays a key role in numerous metabolic pathways such as TCA cycle, gluconeogenesis, glycolysis and fatty acid oxidation. Oxygenation of the gastrointestinal tract including liver and intestine is dynamically regulated by changes in the blood flow and metabolic activity. Cellular adaptation to low oxygen is mediated by the transcription factors HIF-1α and HIF-2α. HIF-1α regulates glycolytic genes whereas HIF-2α is known to primarily regulate genes involved in cell proliferation and iron metabolism. This review focuses on the role of the oxygen sensing signaling in the regulation of hepatic glucose output with an emphasis on hypoxia inducible factor (HIF)-2α. Recent studies have established a metabolic role of HIF-2α in systemic glucose homeostasis. Understanding the HIF-2α dependent mechanism in hepatic metabolism will greatly enhance our potential to utilize the oxygen sensing mechanisms to treat metabolic diseases.
Collapse
Affiliation(s)
- Sadeesh K Ramakrishnan
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.,Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
187
|
Zhao G, Wirth D, Schmitz I, Meyer-Hermann M. A mathematical model of the impact of insulin secretion dynamics on selective hepatic insulin resistance. Nat Commun 2017; 8:1362. [PMID: 29118381 PMCID: PMC5678123 DOI: 10.1038/s41467-017-01627-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 10/03/2017] [Indexed: 12/15/2022] Open
Abstract
Physiological insulin secretion exhibits various temporal patterns, the dysregulation of which is involved in diabetes development. We analyzed the impact of first-phase and pulsatile insulin release on glucose and lipid control with various hepatic insulin signaling networks. The mathematical model suggests that atypical protein kinase C (aPKC) undergoes a bistable switch-on and switch-off, under the control of insulin receptor substrate 2 (IRS2). The activation of IRS1 and IRS2 is temporally separated due to the inhibition of IRS1 by aPKC. The model further shows that the timing of aPKC switch-off is delayed by reduced first-phase insulin and reduced amplitude of insulin pulses. Based on these findings, we propose a sequential model of postprandial hepatic control of glucose and lipid by insulin, according to which delayed aPKC switch-off contributes to selective hepatic insulin resistance, which is a long-standing paradox in the field. Dysregulation of insulin secretion dynamics plays a role in diabetes development. Here, the authors build a mathematical model of hepatic insulin signaling and propose a sequential model of post-meal control of glucose and lipids, according to which delayed aPKC suppression would contribute to selective hepatic insulin resistance.
Collapse
Affiliation(s)
- Gang Zhao
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Rebenring 56, 38106, Braunschweig, Germany
| | - Dagmar Wirth
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Braunschweig, Germany.,Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Ingo Schmitz
- Systems-Oriented Immunology and Inflammation Research Group, Department of Immune Control, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Braunschweig, Germany.,Institute for Molecular and Clinical Immunology, Otto-von-Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Rebenring 56, 38106, Braunschweig, Germany. .,Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Spielmannstraße 7, 38106, Braunschweig, Germany.
| |
Collapse
|
188
|
More VR, Lao J, McLaren DG, Cumiskey AM, Murphy BA, Chen Y, Previs S, Stout S, Patel R, Satapati S, Li W, Kowalik E, Szeto D, Nawrocki A, Pocai A, Wang L, Carrington P. Glucagon like receptor 1/ glucagon dual agonist acutely enhanced hepatic lipid clearance and suppressed de novo lipogenesis in mice. PLoS One 2017; 12:e0186586. [PMID: 29065174 PMCID: PMC5655430 DOI: 10.1371/journal.pone.0186586] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 10/03/2017] [Indexed: 02/08/2023] Open
Abstract
Lipid lowering properties of glucagon have been reported. Blocking glucagon signaling leads to rise in plasma LDL levels. Here, we demonstrate the lipid lowering effects of acute dosing with Glp1r/Gcgr dual agonist (DualAG). All the experiments were performed in 25 week-old male diet-induced (60% kCal fat) obese mice. After 2 hrs of fasting, mice were injected subcutaneously with vehicle, liraglutide (25nmol/kg) and DualAG (25nmol/kg). De novo cholesterol and palmitate synthesis was measured by deuterium incorporation method using D2O. 13C18-oleate infusion was used for measuring fatty acid esterification. Simultaneous activation of Glp1r and Gcgr resulted in decrease in plasma triglyceride and cholesterol levels. DualAG enhanced hepatic LDLr protein levels, along with causing decrease in content of plasma ApoB48 and ApoB100. VLDL secretion, de novo palmitate synthesis and fatty acid esterification decreased with acute DualAG treatment. On the other hand, ketone levels were elevated with DualAG treatment, indicating increased fatty acid oxidation. Lipid relevant changes were absent in liraglutide treated group. In an acute treatment, DualAG demonstrated significant impact on lipid homeostasis, specifically on hepatic uptake, VLDL secretion and de novo synthesis. These effects collectively reveal that lipid lowering abilities of DualAG are primarily through glucagon signaling and are liver centric.
Collapse
Affiliation(s)
- Vijay R. More
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Julie Lao
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - David G. McLaren
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Anne-Marie Cumiskey
- In-Vivo Pharmacology, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Beth Ann Murphy
- In-Vivo Pharmacology, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Ying Chen
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Stephen Previs
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Steven Stout
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Rajesh Patel
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Santhosh Satapati
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Wenyu Li
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Edward Kowalik
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Daphne Szeto
- In-Vivo Pharmacology, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Andrea Nawrocki
- In-Vivo Pharmacology, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Alessandro Pocai
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Liangsu Wang
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Paul Carrington
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| |
Collapse
|
189
|
Quinn WJ, Wan M, Shewale SV, Gelfer R, Rader DJ, Birnbaum MJ, Titchenell PM. mTORC1 stimulates phosphatidylcholine synthesis to promote triglyceride secretion. J Clin Invest 2017; 127:4207-4215. [PMID: 29035283 DOI: 10.1172/jci96036] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/05/2017] [Indexed: 12/13/2022] Open
Abstract
Liver triacylglycerol (TAG) synthesis and secretion are closely linked to nutrient availability. After a meal, hepatic TAG formation from fatty acids is decreased, largely due to a reduction in circulating free fatty acids (FFA). Despite the postprandial decrease in FFA-driven esterification and oxidation, VLDL-TAG secretion is maintained to support peripheral lipid delivery and metabolism. The regulatory mechanisms underlying the postprandial control of VLDL-TAG secretion remain unclear. Here, we demonstrated that the mTOR complex 1 (mTORC1) is essential for this sustained VLDL-TAG secretion and lipid homeostasis. In murine models, the absence of hepatic mTORC1 reduced circulating TAG, despite hepatosteatosis, while activation of mTORC1 depleted liver TAG stores. Additionally, mTORC1 promoted TAG secretion by regulating phosphocholine cytidylyltransferase α (CCTα), the rate-limiting enzyme involved in the synthesis of phosphatidylcholine (PC). Increasing PC synthesis in mice lacking mTORC1 rescued hepatosteatosis and restored TAG secretion. These data identify mTORC1 as a major regulator of phospholipid biosynthesis and subsequent VLDL-TAG secretion, leading to increased postprandial TAG secretion.
Collapse
Affiliation(s)
| | - Min Wan
- Institute for Diabetes, Obesity, and Metabolism, and
| | - Swapnil V Shewale
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Daniel J Rader
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Morris J Birnbaum
- Institute for Diabetes, Obesity, and Metabolism, and.,Internal Medicine, Pfizer Inc., Cambridge, Massachusetts, USA
| | - Paul M Titchenell
- Institute for Diabetes, Obesity, and Metabolism, and.,Department of Physiology Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
190
|
Kandror K. Mammalian target of rapamycin complex 1 and FoxO1 in the transcriptional control of lipolysis and de novo lipogenesis. Curr Opin Endocrinol Diabetes Obes 2017; 24:326-331. [PMID: 28841634 PMCID: PMC11210950 DOI: 10.1097/med.0000000000000352] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE OF REVIEW Postprandial suppression of lipolysis in adipose tissue and stimulation of de novo lipogenesis (DNL) in the liver by insulin are essential for the metabolic homeostasis in the mammalian organism. The mechanism of coregulation of lipolysis and DNL is not clear. RECENT FINDINGS Insulin controls both lipolysis and DNL at the level of transcription via the same mammalian target of rapamycin complex 1 (mTORC1) and FoxO1-mediated signaling pathways. SUMMARY mTORC1 suppresses lipolysis in adipose tissue and activates DNL in the liver, whereas FoxO1 has the opposite effect. Individual inputs of either mTORC1 or FoxO1 in the regulation of lipid metabolism may be difficult to evaluate because of the cross talk between these pathways.
Collapse
Affiliation(s)
- K.V. Kandror
- Boston University School of Medicine, Department of Biochemistry, K123D, 715 Albany Street, Boston, MA 02118, USA. Phone: 617-638-5049
| |
Collapse
|
191
|
Abstract
The liver is crucial for the maintenance of normal glucose homeostasis - it produces glucose during fasting and stores glucose postprandially. However, these hepatic processes are dysregulated in type 1 and type 2 diabetes mellitus, and this imbalance contributes to hyperglycaemia in the fasted and postprandial states. Net hepatic glucose production is the summation of glucose fluxes from gluconeogenesis, glycogenolysis, glycogen synthesis, glycolysis and other pathways. In this Review, we discuss the in vivo regulation of these hepatic glucose fluxes. In particular, we highlight the importance of indirect (extrahepatic) control of hepatic gluconeogenesis and direct (hepatic) control of hepatic glycogen metabolism. We also propose a mechanism for the progression of subclinical hepatic insulin resistance to overt fasting hyperglycaemia in type 2 diabetes mellitus. Insights into the control of hepatic gluconeogenesis by metformin and insulin and into the role of lipid-induced hepatic insulin resistance in modifying gluconeogenic and net hepatic glycogen synthetic flux are also discussed. Finally, we consider the therapeutic potential of strategies that target hepatosteatosis, hyperglucagonaemia and adipose lipolysis.
Collapse
Affiliation(s)
- Max C Petersen
- Department of Internal Medicine, Yale School of Medicine
- Department of Cellular &Molecular Physiology, Yale School of Medicine
| | | | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine
- Department of Cellular &Molecular Physiology, Yale School of Medicine
- Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut 06520, USA
| |
Collapse
|
192
|
Insulin action and resistance in obesity and type 2 diabetes. Nat Med 2017; 23:804-814. [PMID: 28697184 DOI: 10.1038/nm.4350] [Citation(s) in RCA: 841] [Impact Index Per Article: 105.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 05/11/2017] [Indexed: 12/12/2022]
Abstract
Nutritional excess is a major forerunner of type 2 diabetes. It enhances the secretion of insulin, but attenuates insulin's metabolic actions in the liver, skeletal muscle and adipose tissue. However, conflicting evidence indicates a lack of knowledge of the timing of these events during the development of obesity and diabetes, pointing to a key gap in our understanding of metabolic disease. This Perspective reviews alternate viewpoints and recent results on the temporal and mechanistic connections between hyperinsulinemia, obesity and insulin resistance. Although much attention has addressed early steps in the insulin signaling cascade, insulin resistance in obesity seems to be largely elicited downstream of these steps. New findings also connect insulin resistance to extensive metabolic cross-talk between the liver, adipose tissue, pancreas and skeletal muscle. These and other advances over the past 5 years offer exciting opportunities and daunting challenges for the development of new therapeutic strategies for the treatment of type 2 diabetes.
Collapse
|
193
|
Schwarz JM, Noworolski SM, Erkin-Cakmak A, Korn NJ, Wen MJ, Tai VW, Jones GM, Palii SP, Velasco-Alin M, Pan K, Patterson BW, Gugliucci A, Lustig RH, Mulligan K. Effects of Dietary Fructose Restriction on Liver Fat, De Novo Lipogenesis, and Insulin Kinetics in Children With Obesity. Gastroenterology 2017; 153:743-752. [PMID: 28579536 PMCID: PMC5813289 DOI: 10.1053/j.gastro.2017.05.043] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 04/26/2017] [Accepted: 05/21/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Consumption of sugar is associated with obesity, type 2 diabetes mellitus, nonalcoholic fatty liver disease, and cardiovascular disease. The conversion of fructose to fat in liver (de novo lipogenesis [DNL]) may be a modifiable pathogenetic pathway. We determined the effect of 9 days of isocaloric fructose restriction on DNL, liver fat, visceral fat (VAT), subcutaneous fat, and insulin kinetics in obese Latino and African American children with habitual high sugar consumption (fructose intake >50 g/d). METHODS Children (9-18 years old; n = 41) had all meals provided for 9 days with the same energy and macronutrient composition as their standard diet, but with starch substituted for sugar, yielding a final fructose content of 4% of total kilocalories. Metabolic assessments were performed before and after fructose restriction. Liver fat, VAT, and subcutaneous fat were determined by magnetic resonance spectroscopy and imaging. The fractional DNL area under the curve value was measured using stable isotope tracers and gas chromatography/mass spectrometry. Insulin kinetics were calculated from oral glucose tolerance tests. Paired analyses compared change from day 0 to day 10 within each child. RESULTS Compared with baseline, on day 10, liver fat decreased from a median of 7.2% (interquartile range [IQR], 2.5%-14.8%) to 3.8% (IQR, 1.7%-15.5%) (P < .001) and VAT decreased from 123 cm3 (IQR, 85-145 cm3) to 110 cm3 (IQR, 84-134 cm3) (P < .001). The DNL area under the curve decreased from 68% (IQR, 46%-83%) to 26% (IQR, 16%-37%) (P < .001). Insulin kinetics improved (P < .001). These changes occurred irrespective of baseline liver fat. CONCLUSIONS Short-term (9 days) isocaloric fructose restriction decreased liver fat, VAT, and DNL, and improved insulin kinetics in children with obesity. These findings support efforts to reduce sugar consumption. ClinicalTrials.gov Number: NCT01200043.
Collapse
Affiliation(s)
- Jean-Marc Schwarz
- Touro University California College of Osteopathic Medicine, Vallejo, California; Department of Medicine, Division of Endocrinology, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, California.
| | - Susan M Noworolski
- University of California, San Francisco Department of Radiology and Biomedical Imaging, San Francisco, CA, USA
| | - Ayca Erkin-Cakmak
- University of California, San Francisco Department of Pediatrics, UCSF Benioff Children’s Hospital, San Francisco, CA, USA
| | - Natalie J Korn
- University of California, San Francisco Department of Radiology and Biomedical Imaging, San Francisco, CA, USA
| | - Michael J Wen
- University of California, San Francisco Department of Medicine, Division of Endocrinology, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - Viva W Tai
- University of California, San Francisco Clinical and Translational Science Institute Clinical Research Service, San Francisco, CA, USA
| | - Grace M Jones
- Touro University - California College of Osteopathic Medicine, Vallejo, CA, USA
| | - Sergiu P Palii
- Touro University - California College of Osteopathic Medicine, Vallejo, CA, USA
| | - Moises Velasco-Alin
- Touro University - California College of Osteopathic Medicine, Vallejo, CA, USA,University of California, San Francisco Department of Medicine, Division of Endocrinology, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - Karen Pan
- University of California, San Francisco Department of Medicine, Division of Endocrinology, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - Bruce W Patterson
- Washington University School of Medicine, Department of Internal Medicine, St. Louis, MO, USA
| | - Alejandro Gugliucci
- Touro University - California College of Osteopathic Medicine, Vallejo, CA, USA
| | - Robert H Lustig
- University of California, San Francisco Department of Pediatrics, UCSF Benioff Children’s Hospital, San Francisco, CA, USA
| | - Kathleen Mulligan
- Touro University - California College of Osteopathic Medicine, Vallejo, CA, USA,University of California, San Francisco Department of Medicine, Division of Endocrinology, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| |
Collapse
|
194
|
Ghanem SS, Muturi HT, DeAngelis AM, Hu J, Kulkarni RN, Heinrich G, Najjar SM. Age-dependent insulin resistance in male mice with null deletion of the carcinoembryonic antigen-related cell adhesion molecule 2 gene. Diabetologia 2017; 60:1751-1760. [PMID: 28567513 PMCID: PMC5709176 DOI: 10.1007/s00125-017-4307-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/21/2017] [Indexed: 10/19/2022]
Abstract
AIMS/HYPOTHESIS Cc2 -/- mice lacking the gene encoding the carcinoembryonic-antigen-related cell adhesion molecule 2 (Cc2 [also known as Ceacam2]) exhibit hyperphagia that leads to obesity and insulin resistance. This starts at 2 months of age in female mice. Male mutants maintain normal body weight and insulin sensitivity until the last age previously examined (7-8 months), owing to increased sympathetic tone to white adipose tissue and energy expenditure. The current study investigates whether insulin resistance develops in mutant male mice at a later age and whether this is accompanied by changes in insulin homeostasis. METHODS Insulin response was assessed by insulin and glucose tolerance tests. Energy balance was analysed by indirect calorimetry. RESULTS Male Cc2 -/- mice developed overt metabolic abnormalities at about 9 months of age. These include elevated global fat mass, hyperinsulinaemia and insulin resistance (as determined by glucose and insulin intolerance, fed hyperglycaemia and decreased insulin signalling pathways). Pair-feeding experiments showed that insulin resistance resulted from hyperphagia. Indirect calorimetry demonstrated that older mutant male mice had compromised energy expenditure. Despite increased insulin secretion caused by Cc2 deletion, chronic hyperinsulinaemia did not develop in mutant male mice until about 9 months of age, at which point insulin clearance began to decline substantially. This was probably mediated by a marked decrease in hepatic CEACAM1 expression. CONCLUSIONS/INTERPRETATION The data demonstrate that at about 9 months of age, Cc2 -/- male mice develop a reduction in energy expenditure and energy imbalance which, combined with a progressive decrease in CEACAM1-dependent hepatic insulin clearance, causes chronic hyperinsulinaemia and sustained age-dependent insulin resistance. This represents a novel mechanistic underpinning of age-related impairment of hepatic insulin clearance.
Collapse
Affiliation(s)
- Simona S Ghanem
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Harrison T Muturi
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Anthony M DeAngelis
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Jiang Hu
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Rohit N Kulkarni
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Garrett Heinrich
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701-2979, USA
| | - Sonia M Najjar
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701-2979, USA.
| |
Collapse
|
195
|
Sugaya Y, Satoh H. Liver-specific G 0 /G 1 switch gene 2 (G0s2) expression promotes hepatic insulin resistance by exacerbating hepatic steatosis in male Wistar rats. J Diabetes 2017; 9:754-763. [PMID: 27624922 DOI: 10.1111/1753-0407.12482] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/18/2016] [Accepted: 09/08/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Hepatic steatosis is strongly associated with insulin resistance. It has been reported that G0 /G1 switch gene 2 (G0s2) inhibits the lipolytic activity of adipose triglyceride lipase, which is a major lipase in the liver as well as in adipocytes. Moreover, G0s2 protein content is increased in the livers of high-fat diet (HFD)-fed rats. In the present study, we investigated the effect of hepatic G0s2 on insulin sensitivity in male Wistar rats. METHODS Male Wistar rats were fed a 60% HFD for 4 weeks. After 3 weeks of feeding, rats were injected with adenovirus-expressing green fluorescent protein (Ad-GFP; control) or adenovirus-expressing mouse G0s2 (Ad-G0s2). On Day 7 after injection, a euglycemic-hyperinsulinemic clamp study was performed in rats fasted for 8 h. RESULTS Body weight and fasting glucose levels were not significantly different between the Ad-GFP and Ad-G0s2 groups. During the clamp study, the glucose infusion rate required for euglycemia decreased significantly by 16% in the Ad-G0s2 compared with Ad-GFP group. The insulin-suppressed hepatic glucose output increased significantly in the Ad-G0s2 group, but the insulin-stimulated glucose disposal rate was not significantly different between the two groups. Consistent with the clamp data, insulin-stimulated phosphorylation of Akt decreased significantly in livers of rats injected with Ad-G0s2. Furthermore, Oil Red O-staining indicated that overexpression of G0s2 protein in the liver promoted hepatic steatosis by 2.5-fold in HFD-fed rats. CONCLUSION The results of the present study indicate that hepatic G0s2 protein may promote hepatic insulin resistance by exacerbating hepatic steatosis.
Collapse
Affiliation(s)
- Yoshiyuki Sugaya
- Department of Nephrology, Hypertension, Diabetology, Endocrinology, and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Hiroaki Satoh
- Department of Nephrology, Hypertension, Diabetology, Endocrinology, and Metabolism, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
196
|
Leiter SM, Parker VER, Welters A, Knox R, Rocha N, Clark G, Payne F, Lotta L, Harris J, Guerrero-Fernández J, González-Casado I, García-Miñaur S, Gordo G, Wareham N, Martínez-Glez V, Allison M, O’Rahilly S, Barroso I, Meissner T, Davies S, Hussain K, Temple K, Barreda-Bonis AC, Kummer S, Semple RK. Hypoinsulinaemic, hypoketotic hypoglycaemia due to mosaic genetic activation of PI3-kinase. Eur J Endocrinol 2017; 177:175-186. [PMID: 28566443 PMCID: PMC5488397 DOI: 10.1530/eje-17-0132] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/16/2017] [Accepted: 05/30/2017] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Genetic activation of the insulin signal-transducing kinase AKT2 causes syndromic hypoketotic hypoglycaemia without elevated insulin. Mosaic activating mutations in class 1A phospatidylinositol-3-kinase (PI3K), upstream from AKT2 in insulin signalling, are known to cause segmental overgrowth, but the metabolic consequences have not been systematically reported. We assess the metabolic phenotype of 22 patients with mosaic activating mutations affecting PI3K, thereby providing new insight into the metabolic function of this complex node in insulin signal transduction. METHODS Three patients with megalencephaly, diffuse asymmetric overgrowth, hypoketotic, hypoinsulinaemic hypoglycaemia and no AKT2 mutation underwent further genetic, clinical and metabolic investigation. Signalling in dermal fibroblasts from one patient and efficacy of the mTOR inhibitor Sirolimus on pathway activation were examined. Finally, the metabolic profile of a cohort of 19 further patients with mosaic activating mutations in PI3K was assessed. RESULTS In the first three patients, mosaic mutations in PIK3CA (p.Gly118Asp or p.Glu726Lys) or PIK3R2 (p.Gly373Arg) were found. In different tissue samples available from one patient, the PIK3CA p.Glu726Lys mutation was present at burdens from 24% to 42%, with the highest level in the liver. Dermal fibroblasts showed increased basal AKT phosphorylation which was potently suppressed by Sirolimus. Nineteen further patients with mosaic mutations in PIK3CA had neither clinical nor biochemical evidence of hypoglycaemia. CONCLUSIONS Mosaic mutations activating class 1A PI3K cause severe non-ketotic hypoglycaemia in a subset of patients, with the metabolic phenotype presumably related to the extent of mosaicism within the liver. mTOR or PI3K inhibitors offer the prospect for future therapy.
Collapse
Affiliation(s)
- Sarah M Leiter
- Metabolic Research LaboratoriesWellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- The National Institute for Health ResearchCambridge Biomedical Research Centre, Cambridge, UK
| | - Victoria E R Parker
- Metabolic Research LaboratoriesWellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- The National Institute for Health ResearchCambridge Biomedical Research Centre, Cambridge, UK
| | - Alena Welters
- Department of General PaediatricsNeonatology and Paediatric Cardiology, University Children’s Hospital, Düsseldorf, Germany
| | - Rachel Knox
- Metabolic Research LaboratoriesWellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- The National Institute for Health ResearchCambridge Biomedical Research Centre, Cambridge, UK
| | - Nuno Rocha
- Metabolic Research LaboratoriesWellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- The National Institute for Health ResearchCambridge Biomedical Research Centre, Cambridge, UK
| | - Graeme Clark
- Department of Molecular GeneticsAddenbrooke’s Hospital, Cambridge, UK
| | | | - Luca Lotta
- MRC Epidemiology UnitUniversity of Cambridge, Cambridge, UK
| | - Julie Harris
- Metabolic Research LaboratoriesWellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- The National Institute for Health ResearchCambridge Biomedical Research Centre, Cambridge, UK
| | | | | | - Sixto García-Miñaur
- Departments of Clinical and Molecular GeneticsLa Paz Hospital, Madrid, Spain
| | - Gema Gordo
- Departments of Clinical and Molecular GeneticsLa Paz Hospital, Madrid, Spain
| | - Nick Wareham
- MRC Epidemiology UnitUniversity of Cambridge, Cambridge, UK
| | | | | | - Stephen O’Rahilly
- Metabolic Research LaboratoriesWellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- The National Institute for Health ResearchCambridge Biomedical Research Centre, Cambridge, UK
| | - Inês Barroso
- Metabolic Research LaboratoriesWellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- The National Institute for Health ResearchCambridge Biomedical Research Centre, Cambridge, UK
- Wellcome Trust Sanger InstituteHinxton, Cambridge, UK
| | - Thomas Meissner
- Department of General PaediatricsNeonatology and Paediatric Cardiology, University Children’s Hospital, Düsseldorf, Germany
| | - Susan Davies
- Departments of HistopathologyAddenbrooke’s Hospital, Cambridge, UK
| | - Khalid Hussain
- Institute of Child HealthUniversity College London, London, UK
| | - Karen Temple
- Department of Clinical GeneticsUniversity Hospital Southampton, Southampton, UK
| | | | - Sebastian Kummer
- Department of General PaediatricsNeonatology and Paediatric Cardiology, University Children’s Hospital, Düsseldorf, Germany
| | - Robert K Semple
- Metabolic Research LaboratoriesWellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- The National Institute for Health ResearchCambridge Biomedical Research Centre, Cambridge, UK
| |
Collapse
|
197
|
Caputo T, Gilardi F, Desvergne B. From chronic overnutrition to metaflammation and insulin resistance: adipose tissue and liver contributions. FEBS Lett 2017; 591:3061-3088. [DOI: 10.1002/1873-3468.12742] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/28/2017] [Accepted: 07/02/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Tiziana Caputo
- Center for Integrative Genomics; Lausanne Faculty of Biology and Medicine; University of Lausanne; Switzerland
| | - Federica Gilardi
- Center for Integrative Genomics; Lausanne Faculty of Biology and Medicine; University of Lausanne; Switzerland
| | - Béatrice Desvergne
- Center for Integrative Genomics; Lausanne Faculty of Biology and Medicine; University of Lausanne; Switzerland
| |
Collapse
|
198
|
Bergman RN, Iyer MS. Indirect Regulation of Endogenous Glucose Production by Insulin: The Single Gateway Hypothesis Revisited. Diabetes 2017. [PMID: 28637826 DOI: 10.2337/db16-1320] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
On the basis of studies that investigated the intraportal versus systemic insulin infusion and transendothelial transport of insulin, we proposed the "single gateway hypothesis," which supposes an indirect regulation of hepatic glucose production by insulin; the rate-limiting transport of insulin across the adipose tissue capillaries is responsible for the slow suppression of free fatty acids (FFAs), which in turn is responsible for delayed suppression of hepatic endogenous glucose production (EGP) during insulin infusion. Preventing the fall in plasma FFAs during insulin infusion either by administering intralipids or by inhibiting adipose tissue lipolysis led to failure in EGP suppression, thus supporting our hypothesis. More recently, mice lacking hepatic Foxo1 in addition to Akt1 and Akt2 (L-AktFoxo1TKO), all required for insulin signaling, surprisingly showed normal glycemia. Inhibiting the fall of plasma FFAs in these mice prevented the suppression of EGP during a clamp, reaffirming that the site of insulin action to control EGP is extrahepatic. Measuring whole-body turnover rates of glucose and FFAs in L-AktFoxo1TKO mice also confirmed that hepatic EGP was regulated by insulin-mediated control of FFAs. The knockout mouse model in combination with sophisticated molecular techniques confirmed our physiological findings and the single gateway hypothesis.
Collapse
Affiliation(s)
- Richard N Bergman
- Cedars-Sinai Diabetes and Obesity Research Institute, Los Angeles, CA
| | - Malini S Iyer
- Cedars-Sinai Diabetes and Obesity Research Institute, Los Angeles, CA
| |
Collapse
|
199
|
Titchenell PM, Lazar MA, Birnbaum MJ. Unraveling the Regulation of Hepatic Metabolism by Insulin. Trends Endocrinol Metab 2017; 28:497-505. [PMID: 28416361 PMCID: PMC5477655 DOI: 10.1016/j.tem.2017.03.003] [Citation(s) in RCA: 296] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/15/2017] [Accepted: 03/23/2017] [Indexed: 01/26/2023]
Abstract
During insulin-resistant states such as type 2 diabetes mellitus (T2DM), insulin fails to suppress hepatic glucose production but promotes lipid synthesis leading to hyperglycemia and hypertriglyceridemia. Defining the downstream signaling pathways underlying the control of hepatic metabolism by insulin is necessary for understanding both normal physiology and the pathogenesis of metabolic disease. We summarize recent literature highlighting the importance of both hepatic and extrahepatic mechanisms in insulin regulation of liver glucose and lipid metabolism. We posit that a failure of insulin to inappropriately regulate liver metabolism during T2DM is not exclusively from an inherent defect in canonical liver insulin signaling but is instead due to a combination of hyperinsulinemia, altered substrate supply, and the input of several extrahepatic signals.
Collapse
Affiliation(s)
- Paul M Titchenell
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Morris J Birnbaum
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Present address: Internal Medicine, Pfizer Inc., Cambridge, MA, USA.
| |
Collapse
|
200
|
Abstract
Adipose tissue represents a critical component in healthy energy homeostasis. It fulfills important roles in whole-body lipid handling, serves as the body's major energy storage compartment and insulation barrier, and secretes numerous endocrine mediators such as adipokines or lipokines. As a consequence, dysfunction of these processes in adipose tissue compartments is tightly linked to severe metabolic disorders, including obesity, metabolic syndrome, lipodystrophy, and cachexia. While numerous studies have addressed causes and consequences of obesity-related adipose tissue hypertrophy and hyperplasia for health, critical pathways and mechanisms in (involuntary) adipose tissue loss as well as its systemic metabolic consequences are far less understood. In this review, we discuss the current understanding of conditions of adipose tissue wasting and review microenvironmental determinants of adipocyte (dys)function in related pathophysiologies.
Collapse
Affiliation(s)
- Alexandros Vegiopoulos
- Junior Group Metabolism and Stem Cell Plasticity, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Maria Rohm
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Joint Heidelberg-IDC Translational Diabetes Program Inner Medicine I, Neuherberg, Germany
| |
Collapse
|