1
|
González JT, Scharfman OH, Zhu W, Kasamoto J, Gould V, Perry RJ, Higgins-Chen AT. Transcriptomic and epigenomic signatures of liver metabolism and insulin sensitivity in aging mice. Mech Ageing Dev 2025; 225:112068. [PMID: 40324540 DOI: 10.1016/j.mad.2025.112068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/25/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Age-related declines in insulin sensitivity and glucose metabolism contribute to metabolic disease. Despite the liver's central role in glucose homeostasis, a comprehensive phenotypic characterization and concurrent molecular analysis of insulin resistance and metabolic dysfunction in the aging liver is lacking. We characterized hepatic insulin resistance and mitochondrial metabolic defects through metabolic cage, hyperinsulinemic-euglycemic clamp, and tracer studies paired with transcriptomic and DNA methylation analyses in young and aged male mice. Aged mice exhibited benchmark measures of whole body and liver insulin resistance. Aged mice showed lower pyruvate dehydrogenase flux, decreased fatty acid oxidation and citrate synthase fluxes, and increased pyruvate carboxylase flux under insulin-stimulated conditions. Molecular analysis revealed age-related changes in metabolic genes Pck1, Socs3, Tbc1d4, and Enpp1. Unsupervised network analysis identified an intercorrelated phenotype module (ME-Glucose), RNA module, and DNA methylation module. The DNA methylation module was enriched for lipid metabolism pathways and TCF-1 binding, while the RNA module was enriched for MZF-1 binding and regulation by miR-155-5p. Protein-protein interaction network analysis revealed interactions between module genes and canonical metabolic pathways, highlighting genes including Ets1, Ppp1r3b, and Enpp3. This study reveals novel genes underlying age-related hepatic insulin resistance as potential targets for metabolic interventions to promote healthy aging.
Collapse
Affiliation(s)
- John T González
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Olivia H Scharfman
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Wanling Zhu
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA; Department of Endocrinology & Metabolism, Yale School of Medicine, New Haven, CT, USA
| | - Jessica Kasamoto
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Victoria Gould
- Altos Labs, Institute of Computation, San Diego, CA 92114, USA
| | - Rachel J Perry
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA; Department of Endocrinology & Metabolism, Yale School of Medicine, New Haven, CT, USA.
| | - Albert T Higgins-Chen
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA; Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
2
|
Rahim M, Bednarski TK, Hasenour CM, Banerjee DR, Trenary I, Young JD. Simultaneous in vivo multi-organ fluxomics reveals divergent metabolic adaptations in liver, heart, and skeletal muscle during obesity. Cell Rep 2025; 44:115591. [PMID: 40244853 PMCID: PMC12167616 DOI: 10.1016/j.celrep.2025.115591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 02/23/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
We present an isotope-based metabolic flux analysis (MFA) approach to simultaneously quantify metabolic fluxes in the liver, heart, and skeletal muscle of individual mice. The platform was scaled to examine metabolic flux adaptations in age-matched cohorts of mice exhibiting varying levels of chronic obesity. We found that severe obesity increases hepatic gluconeogenesis and citric acid cycle flux, accompanied by elevated glucose oxidation in the heart that compensates for impaired fatty acid oxidation. In contrast, skeletal muscle fluxes exhibit an overall reduction in substrate oxidation. These findings demonstrate the dichotomy in fuel utilization between cardiac and skeletal muscle during worsening metabolic disease and demonstrate the divergent effects of obesity on metabolic fluxes in different organs. This multi-tissue MFA technology can be extended to address important questions about in vivo regulation of metabolism and its dysregulation in disease, which cannot be fully answered through studies of single organs or isolated cells/tissues.
Collapse
Affiliation(s)
- Mohsin Rahim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Tomasz K Bednarski
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Clinton M Hasenour
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Deveena R Banerjee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Irina Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
3
|
Hemalatha A, Li Z, Gonzalez DG, Matte-Martone C, Tai K, Lathrop E, Gil D, Ganesan S, Gonzalez LE, Skala M, Perry RJ, Greco V. Metabolic rewiring in skin epidermis drives tolerance to oncogenic mutations. Nat Cell Biol 2025; 27:218-231. [PMID: 39762578 PMCID: PMC11821535 DOI: 10.1038/s41556-024-01574-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 11/01/2024] [Indexed: 02/06/2025]
Abstract
Skin epithelial stem cells correct aberrancies induced by oncogenic mutations. Oncogenes invoke different strategies of epithelial tolerance; while wild-type cells outcompete β-catenin-gain-of-function (βcatGOF) cells, HrasG12V cells outcompete wild-type cells. Here we ask how metabolic states change as wild-type stem cells interface with mutant cells and drive different cell-competition outcomes. By tracking the endogenous redox ratio (NAD(P)H/FAD) with single-cell resolution in the same mouse over time, we discover that βcatGOF and HrasG12V mutations, when interfaced with wild-type epidermal stem cells, lead to a rapid drop in redox ratios, indicating more oxidized cellular redox. However, the resultant redox differential persists through time in βcatGOF, whereas it is flattened rapidly in the HrasG12Vmodel. Using 13C liquid chromatography-tandem mass spectrometry, we find that the βcatGOF and HrasG12V mutant epidermis increase the fractional contribution of glucose through the oxidative tricarboxylic acid cycle. Treatment with metformin, a modifier of cytosolic redox, inhibits downstream mutant phenotypes and reverses cell-competition outcomes of both mutant models.
Collapse
Affiliation(s)
| | - Zongyu Li
- Departments of Cellular & Molecular Physiology and Internal Medicine (Endocrinology), Yale School of Medicine, New Haven, CT, USA
| | - David G Gonzalez
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | | | - Karen Tai
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | | | - Daniel Gil
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - Smirthy Ganesan
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | | | - Melissa Skala
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - Rachel J Perry
- Departments of Cellular & Molecular Physiology and Internal Medicine (Endocrinology), Yale School of Medicine, New Haven, CT, USA.
| | - Valentina Greco
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.
- Departments of Cell Biology and Dermatology, Yale Stem Cell Center, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.
- Howard Hughes Medical Institute (HHMI), Chevy Chase, MD, USA.
| |
Collapse
|
4
|
Pesta D, Anadol-Schmitz E, Sarabhai T, Op den Kamp Y, Gancheva S, Trinks N, Zaharia OP, Mastrototaro L, Lyu K, Habets I, Op den Kamp-Bruls YMH, Dewidar B, Weiss J, Schrauwen-Hinderling V, Zhang D, Gaspar RC, Strassburger K, Kupriyanova Y, Al-Hasani H, Szendroedi J, Schrauwen P, Phielix E, Shulman GI, Roden M. Determinants of increased muscle insulin sensitivity of exercise-trained versus sedentary normal weight and overweight individuals. SCIENCE ADVANCES 2025; 11:eadr8849. [PMID: 39742483 PMCID: PMC11691647 DOI: 10.1126/sciadv.adr8849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/26/2024] [Indexed: 01/03/2025]
Abstract
The athlete's paradox states that intramyocellular triglyceride accumulation associates with insulin resistance in sedentary but not in endurance-trained humans. Underlying mechanisms and the role of muscle lipid distribution and composition on glucose metabolism remain unclear. We compared highly trained athletes (ATHL) with sedentary normal weight (LEAN) and overweight-to-obese (OVWE) male and female individuals. This observational study found that ATHL show higher insulin sensitivity, muscle mitochondrial content, and capacity, but lower activation of novel protein kinase C (nPKC) isoforms, despite higher diacylglycerol concentrations. Notably, sedentary but insulin sensitive OVWE feature lower plasma membrane-to-mitochondria sn-1,2-diacylglycerol ratios. In ATHL, calpain-2, which cleaves nPKC, negatively associates with PKCε activation and positively with insulin sensitivity along with higher GLUT4 and hexokinase II content. These findings contribute to explaining the athletes' paradox by demonstrating lower nPKC activation, increased calpain, and mitochondrial partitioning of bioactive diacylglycerols, the latter further identifying an obesity subtype with increased insulin sensitivity (NCT03314714).
Collapse
Affiliation(s)
- Dominik Pesta
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Centre for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Evrim Anadol-Schmitz
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Theresia Sarabhai
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Yvo Op den Kamp
- Department of Nutrition and Movement Sciences, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Sofiya Gancheva
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Nina Trinks
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Oana-Patricia Zaharia
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Lucia Mastrototaro
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Kun Lyu
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Ivo Habets
- Department of Nutrition and Movement Sciences, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Yvonne M. H. Op den Kamp-Bruls
- Department of Nutrition and Movement Sciences, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Bedair Dewidar
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Jürgen Weiss
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Vera Schrauwen-Hinderling
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Nutrition and Movement Sciences, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Dongyan Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Klaus Strassburger
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Yuliya Kupriyanova
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Hadi Al-Hasani
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology, Diabetology and Clinical Chemistry (Internal Medicine 1), Heidelberg University Hospital, Heidelberg, Germany
| | - Patrick Schrauwen
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- Leiden University Medical Center, Clinical Epidemiology, Leiden, Netherlands
| | - Esther Phielix
- Department of Nutrition and Movement Sciences, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Gerald I. Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
5
|
Schleh MW, Ryan BJ, Ahn C, Ludzki AC, Van Pelt DW, Pitchford LM, Chugh OK, Luker AT, Luker KE, Samovski D, Abumrad NA, Burant CF, Horowitz JF. Impaired suppression of fatty acid release by insulin is a strong predictor of reduced whole-body insulin-mediated glucose uptake and skeletal muscle insulin receptor activation. Acta Physiol (Oxf) 2025; 241:e14249. [PMID: 39487600 DOI: 10.1111/apha.14249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/06/2024] [Accepted: 09/24/2024] [Indexed: 11/04/2024]
Abstract
AIM To examine factors underlying why most, but not all, adults with obesity exhibit impaired insulin-mediated glucose uptake, we compared: (1) adipose tissue fatty acid (FA) release, (2) skeletal muscle lipid droplet (LD) characteristics, and (3) insulin signalling events, in skeletal muscle of adults with obesity with relatively high versus low insulin-mediated glucose uptake. METHODS Seventeen adults with obesity (BMI: 36 ± 3 kg/m2) completed a 2 h hyperinsulinemic-euglycemic clamp with stable isotope tracer infusions to measure glucose rate of disappearance (glucose Rd) and FA rate of appearance (FA Ra). Skeletal muscle biopsies were collected at baseline and 30 min into the insulin infusion. Participants were stratified into HIGH (n = 7) and LOW (n = 10) insulin sensitivity cohorts by their glucose Rd during the hyperinsulinemic clamp (LOW< 400; HIGH >550 nmol/kgFFM/min/[μU/mL]). RESULTS Insulin-mediated suppression of FA Ra was lower in LOW compared with HIGH (p < 0.01). In skeletal muscle, total intramyocellular lipid content did not differ between cohorts. However, the size of LDs in the subsarcolemmal region (SS) of type II muscle fibres was larger in LOW compared with HIGH (p = 0.01). Additionally, insulin receptor-β (IRβ) interactions with regulatory proteins CD36 and Fyn were lower in LOW versus HIGH (p < 0.01), which aligned with attenuated insulin-mediated Tyr phosphorylation of IRβ and downstream insulin-signalling proteins in LOW. CONCLUSION Collectively, reduced ability for insulin to suppress FA mobilization, with accompanying modifications in intramyocellular LD size and distribution, and diminished IRβ interaction with key regulatory proteins may be key contributors to impaired insulin-mediated glucose uptake commonly found in adults with obesity.
Collapse
Affiliation(s)
- Michael W Schleh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Benjamin J Ryan
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Cheehoon Ahn
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Alison C Ludzki
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Douglas W Van Pelt
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lisa M Pitchford
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Olivia K Chugh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Austin T Luker
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Kathryn E Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Dmitri Samovski
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nada A Abumrad
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Charles F Burant
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jeffrey F Horowitz
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
6
|
Goedeke L, Ma Y, Gaspar RC, Nasiri A, Lee J, Zhang D, Galsgaard KD, Hu X, Zhang J, Guerrera N, Li X, LaMoia T, Hubbard BT, Haedersdal S, Wu X, Stack J, Dufour S, Butrico GM, Kahn M, Perry RJ, Cline GW, Young LH, Shulman GI. SGLT2 inhibition alters substrate utilization and mitochondrial redox in healthy and failing rat hearts. J Clin Invest 2024; 134:e176708. [PMID: 39680452 PMCID: PMC11645152 DOI: 10.1172/jci176708] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 10/17/2024] [Indexed: 12/18/2024] Open
Abstract
Previous studies highlight the potential for sodium-glucose cotransporter type 2 (SGLT2) inhibitors (SGLT2i) to exert cardioprotective effects in heart failure by increasing plasma ketones and shifting myocardial fuel utilization toward ketone oxidation. However, SGLT2i have multiple in vivo effects and the differential impact of SGLT2i treatment and ketone supplementation on cardiac metabolism remains unclear. Here, using gas chromatography-mass spectrometry (GC-MS) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) methodology combined with infusions of [13C6]glucose or [13C4]βOHB, we demonstrate that acute SGLT2 inhibition with dapagliflozin shifts relative rates of myocardial mitochondrial metabolism toward ketone oxidation, decreasing pyruvate oxidation with little effect on fatty acid oxidation in awake rats. Shifts in myocardial ketone oxidation persisted when plasma glucose levels were maintained. In contrast, acute βOHB infusion similarly augmented ketone oxidation, but markedly reduced fatty acid oxidation and did not alter glucose uptake or pyruvate oxidation. After inducing heart failure, dapagliflozin increased relative rates of ketone and fatty acid oxidation, but decreased pyruvate oxidation. Dapagliflozin increased mitochondrial redox and reduced myocardial oxidative stress in heart failure, which was associated with improvements in left ventricular ejection fraction after 3 weeks of treatment. Thus, SGLT2i have pleiotropic effects on systemic and heart metabolism, which are distinct from ketone supplementation and may contribute to the long-term cardioprotective benefits of SGLT2i.
Collapse
Affiliation(s)
- Leigh Goedeke
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Medicine (Cardiology) and The Cardiovascular Research Institute and
- Department of Medicine (Endocrinology) and The Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yina Ma
- Department of Internal Medicine (Cardiovascular Medicine) and The Yale Cardiovascular Research Center, Yale School of Medicine, New Haven Connecticut, USA
| | - Rafael C. Gaspar
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Ali Nasiri
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Jieun Lee
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Dongyan Zhang
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Katrine Douglas Galsgaard
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Xiaoyue Hu
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Jiasheng Zhang
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Nicole Guerrera
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Xiruo Li
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Traci LaMoia
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Brandon T. Hubbard
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Sofie Haedersdal
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Clinical Research, Copenhagen University Hospital, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Xiaohong Wu
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - John Stack
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Sylvie Dufour
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Gina Marie Butrico
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Mario Kahn
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Rachel J. Perry
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Gary W. Cline
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Lawrence H. Young
- Department of Internal Medicine (Cardiovascular Medicine) and The Yale Cardiovascular Research Center, Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Gerald I. Shulman
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|
7
|
Fiorenza M, Onslev J, Henríquez-Olguín C, Persson KW, Hesselager SA, Jensen TE, Wojtaszewski JFP, Hostrup M, Bangsbo J. Reducing the mitochondrial oxidative burden alleviates lipid-induced muscle insulin resistance in humans. SCIENCE ADVANCES 2024; 10:eadq4461. [PMID: 39475607 PMCID: PMC11524190 DOI: 10.1126/sciadv.adq4461] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
Preclinical models suggest mitochondria-derived oxidative stress as an underlying cause of insulin resistance. However, it remains unknown whether this pathophysiological mechanism is conserved in humans. Here, we used an invasive in vivo mechanistic approach to interrogate muscle insulin action while selectively manipulating the mitochondrial redox state in humans. To this end, we conducted insulin clamp studies combining intravenous infusion of a lipid overload with intake of a mitochondria-targeted antioxidant (mitoquinone). Under lipid overload, selective modulation of mitochondrial redox state by mitoquinone enhanced insulin-stimulated glucose uptake in skeletal muscle. Mechanistically, mitoquinone did not affect canonical insulin signaling but augmented insulin-stimulated glucose transporter type 4 (GLUT4) translocation while reducing the mitochondrial oxidative burden under lipid oversupply. Complementary ex vivo studies in human muscle fibers exposed to high intracellular lipid levels revealed that mitoquinone improves features of mitochondrial bioenergetics, including diminished mitochondrial H2O2 emission. These findings provide translational and mechanistic evidence implicating mitochondrial oxidants in the development of lipid-induced muscle insulin resistance in humans.
Collapse
Affiliation(s)
- Matteo Fiorenza
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Johan Onslev
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Carlos Henríquez-Olguín
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
- Exercise Science Laboratory, Faculty of Medicine, Universidad Finis Terrae, Santiago 1509, Chile
| | - Kaspar W. Persson
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Sofie A. Hesselager
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Thomas E. Jensen
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jørgen F. P. Wojtaszewski
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Morten Hostrup
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jens Bangsbo
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| |
Collapse
|
8
|
Gaspar RC, Sakuma I, Nasiri A, Hubbard BT, LaMoia TE, Leitner BP, Tep S, Xi Y, Green EM, Ullman JC, Petersen KF, Shulman GI. Small molecule inhibition of glycogen synthase I reduces muscle glycogen content and improves biomarkers in a mouse model of Pompe disease. Am J Physiol Endocrinol Metab 2024; 327:E524-E532. [PMID: 39171753 PMCID: PMC11482269 DOI: 10.1152/ajpendo.00175.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024]
Abstract
Pompe disease is a rare genetic disorder caused by a deficiency of the enzyme acid alpha-glucosidase (GAA). This enzyme is responsible for breaking down glycogen, leading to the abnormal accumulation of glycogen, which results in progressive muscle weakness and metabolic dysregulation. In this study, we investigated the hypothesis that the small molecule inhibition of glycogen synthase I (GYS1) may reduce muscle glycogen content and improve metabolic dysregulation in a mouse model of Pompe disease. To address this hypothesis, we studied four groups of male mice: a control group of wild-type (WT) B6129SF1/J mice fed either regular chow or a GYS1 inhibitor (MZ-101) diet (WT-GYS1), and Pompe model mice B6;129-Gaatm1Rabn/J fed either regular chow (GAA-KO) or MZ-101 diet (GAA-GYS1) for 7 days. Our findings revealed that GAA-KO mice exhibited abnormal glycogen accumulation in the gastrocnemius, heart, and diaphragm. In contrast, inhibiting GYS1 reduced glycogen levels in all tissues compared with GAA-KO mice. Furthermore, GAA-KO mice displayed reduced spontaneous activity during the dark cycle compared with WT mice, whereas GYS1 inhibition counteracted this effect. Compared with GAA-KO mice, GAA-GYS1 mice exhibited improved glucose tolerance and whole body insulin sensitivity. These improvements in insulin sensitivity could be attributed to increased AMP-activated protein kinase phosphorylation in the gastrocnemius of WT-GYS1 and GAA-GYS1 mice. Additionally, the GYS1 inhibitor led to a reduction in the phosphorylation of GSS641 and the LC3 autophagy marker. Together, our results suggest that targeting GYS1 could serve as a potential strategy for treating glycogen storage disorders and metabolic dysregulation.NEW & NOTEWORTHY We investigated the effects of small molecule inhibition of glycogen synthase I (GYS1) on glucose metabolism in a mouse model of Pompe disease. GYS1 inhibition reduces abnormal glycogen accumulation and molecular biomarkers associated with Pompe disease while also improving glucose intolerance. Our results collectively demonstrate that the GYS1 inhibitor represents a novel approach to substrate reduction therapy for Pompe disease.
Collapse
Affiliation(s)
- Rafael Calais Gaspar
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, United States
| | - Ikki Sakuma
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, United States
| | - Ali Nasiri
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, United States
| | - Brandon T Hubbard
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, United States
| | - Traci E LaMoia
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, United States
| | - Brooks P Leitner
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, United States
| | - Samnang Tep
- Maze Therapeutics, South San Francisco, California,United States
| | - Yannan Xi
- Maze Therapeutics, South San Francisco, California,United States
| | - Eric M Green
- Maze Therapeutics, South San Francisco, California,United States
| | - Julie C Ullman
- Maze Therapeutics, South San Francisco, California,United States
| | - Kitt Falk Petersen
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, United States
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, United States
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States
| |
Collapse
|
9
|
von Rauchhaupt E, Rodemer C, Kliemank E, Bulkescher R, Campos M, Kopf S, Fleming T, Herzig S, Nawroth PP, Szendroedi J, Zemva J, Sulaj A. Glucose Load Following Prolonged Fasting Increases Oxidative Stress- Linked Response in Individuals With Diabetic Complications. Diabetes Care 2024; 47:1584-1592. [PMID: 38905209 PMCID: PMC11362116 DOI: 10.2337/dc24-0209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/28/2024] [Indexed: 06/23/2024]
Abstract
OBJECTIVE Prolonged catabolic states in type 2 diabetes (T2D), exacerbated by excess substrate flux and hyperglycemia, can challenge metabolic flexibility and antioxidative capacity. We investigated cellular responses to glucose load after prolonged fasting in T2D. RESEARCH DESIGN AND METHODS Glucose-tolerant individuals (CON, n = 10) and individuals with T2D with (T2D+, n = 10) and without (T2D-, n = 10) diabetes complications underwent oral glucose tolerance test before and after a 5-day fasting-mimicking diet. Peripheral blood mononuclear cell (PBMC) resistance to ex vivo dicarbonyl methylglyoxal (MG) exposure after glucose load was assessed. Markers of dicarbonyl detoxification, oxidative stress, and mitochondrial biogenesis were analyzed by quantitative PCR, with mitochondrial complex protein expression assessed by Western blotting. RESULTS T2D+ exhibited decreased PBMC resistance against MG, while T2D- resistance remained unchanged, and CON improved postglucose load and fasting (-19.0% vs. -1.7% vs. 12.6%; all P = 0.017). T2D+ showed increased expression in dicarbonyl detoxification (mRNA glyoxalase-1, all P = 0.039), oxidative stress (mRNA glutathione-disulfide-reductase, all P = 0.006), and mitochondrial complex V protein (all P = 0.004) compared with T2D- and CON postglucose load and fasting. Citrate synthase activity remained unchanged, indicating no change in mitochondrial number. Mitochondrial biogenesis increased in T2D- compared with CON postglucose load and fasting (mRNA HspA9, P = 0.032). T2D-, compared with CON, exhibited increased oxidative stress postfasting, but not postglucose load, with increased mRNA expression in antioxidant defenses (mRNA forkhead box O4, P = 0.036, and glutathione-peroxidase-2, P = 0.034), and compared with T2D+ (glutathione-peroxidase-2, P = 0.04). CONCLUSIONS These findings suggest increased susceptibility to glucose-induced oxidative stress in individuals with diabetes complications after prolonged fasting and might help in diet interventions for diabetes management.
Collapse
Affiliation(s)
- Ekaterina von Rauchhaupt
- Department of Endocrinology, Diabetology, Metabolic Diseases and Clinical Chemistry (Internal Medicine I), University Hospital of Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Claus Rodemer
- Department of Endocrinology, Diabetology, Metabolic Diseases and Clinical Chemistry (Internal Medicine I), University Hospital of Heidelberg, Heidelberg, Germany
| | - Elisabeth Kliemank
- Department of Endocrinology, Diabetology, Metabolic Diseases and Clinical Chemistry (Internal Medicine I), University Hospital of Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Ruben Bulkescher
- Department of Endocrinology, Diabetology, Metabolic Diseases and Clinical Chemistry (Internal Medicine I), University Hospital of Heidelberg, Heidelberg, Germany
| | - Marta Campos
- Department of Endocrinology, Diabetology, Metabolic Diseases and Clinical Chemistry (Internal Medicine I), University Hospital of Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Stefan Kopf
- Department of Endocrinology, Diabetology, Metabolic Diseases and Clinical Chemistry (Internal Medicine I), University Hospital of Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Thomas Fleming
- Department of Endocrinology, Diabetology, Metabolic Diseases and Clinical Chemistry (Internal Medicine I), University Hospital of Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Stephan Herzig
- German Center for Diabetes Research, München-Neuherberg, Germany
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, Heidelberg University Hospital, Heidelberg, Germany
- Chair Molecular Metabolic Control, Technical University Munich, Munich, Germany
| | - Peter P. Nawroth
- Department of Endocrinology, Diabetology, Metabolic Diseases and Clinical Chemistry (Internal Medicine I), University Hospital of Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Center Munich, Neuherberg, Germany
| | - Julia Szendroedi
- Department of Endocrinology, Diabetology, Metabolic Diseases and Clinical Chemistry (Internal Medicine I), University Hospital of Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Center Munich, Neuherberg, Germany
| | - Johanna Zemva
- Department of Endocrinology, Diabetology, Metabolic Diseases and Clinical Chemistry (Internal Medicine I), University Hospital of Heidelberg, Heidelberg, Germany
- Joint Practice for Endocrinology, Diabetology and Nuclear Medicine Heidelberg, Heidelberg, Germany
| | - Alba Sulaj
- Department of Endocrinology, Diabetology, Metabolic Diseases and Clinical Chemistry (Internal Medicine I), University Hospital of Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| |
Collapse
|
10
|
Toksoy Z, Ma Y, Goedeke L, Li W, Hu X, Wu X, Cacheux M, Liu Y, Akar FG, Shulman GI, Young LH. Role of AMPK in Atrial Metabolic Homeostasis and Substrate Preference. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.29.608789. [PMID: 39257756 PMCID: PMC11383699 DOI: 10.1101/2024.08.29.608789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Atrial fibrillation is the most common clinical arrhythmia and may be due in part to metabolic stress. Atrial specific deletion of the master metabolic sensor, AMP-activated protein kinase (AMPK), induces atrial remodeling culminating in atrial fibrillation in mice, implicating AMPK signaling in the maintenance of atrial electrical and structural homeostasis. However, atrial substrate preference for mitochondrial oxidation and the role of AMPK in regulating atrial metabolism are unknown. Here, using LC-MS/MS methodology combined with infusions of [ 13 C 6 ]glucose and [ 13 C 4 ]β-hydroxybutyrate in conscious mice, we demonstrate that conditional deletion of atrial AMPK catalytic subunits shifts mitochondrial atrial metabolism away from fatty acid oxidation and towards pyruvate oxidation. LC-MS/MS-based quantification of acyl-CoAs demonstrated decreased atrial tissue content of long-chain fatty acyl-CoAs. Proteomic analysis revealed a broad downregulation of proteins responsible for fatty acid uptake (LPL, CD36, FABP3), acylation and oxidation. Atrial AMPK deletion reduced expression of atrial PGC1-α and downstream PGC1-α/PPARα/RXR regulated gene transcripts. In contrast, atrial [ 14 C]2-deoxyglucose uptake and GLUT1 expression increased with fasting in mice with AMPK deletion, while the expression of glycolytic enzymes exhibited heterogenous changes. Thus, these results highlight the crucial homeostatic role of AMPK in the atrium, with loss of atrial AMPK leading to downregulation of the PGC1-α/PPARα pathway and broad metabolic reprogramming with a loss of fatty acid oxidation, which may contribute to atrial remodeling and arrhythmia.
Collapse
|
11
|
Zhao Y, Qiao M, Wang X, Luo X, Yang J, Hu J. Allantoin reduces glucotoxicity and lipotoxicity in a type 2 diabetes rat model by modulating the PI3K and MAPK signaling pathways. Heliyon 2024; 10:e34716. [PMID: 39144993 PMCID: PMC11320158 DOI: 10.1016/j.heliyon.2024.e34716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
Objective The current study aimed to investigate the potential therapeutic impact of allantoin on diabetes produced by a high-fat diet (HFD) and streptozotocin (STZ) in rats. Subjects and methods Male Sprague-Dawley rats were fed a high-fat diet to induce insulin resistance, followed by streptozotocin injection to induce diabetes. The effect of oral treatment of allantoin (200, 400 and 800 mg/kg/day) for 8 weeks was evaluated by calculating the alteration in metabolic parameters, biochemical indicators, the oral glucose tolerance tests (OGTT) and hyperinsulinemic-euglycemic clamp tests were performed. Histopathological studies were performed in the liver, kidney and pancreas. Next, the expressions of the MAPK and insulin signaling pathway were measured by Western blot analysis to elucidate the potential mechanism underlying these antidiabetic activities. Results The administration of allantoin resulted in a significant decrease in fasting blood glucose (FBG) levels, glycogen levels, and glycosylated hemoglobin levels in diabetic rats. Additionally, allantoin therapy led to a dose-dependent increase in body weight growth and serum insulin levels. In addition, the administration of allantoin resulted in a considerable reduction in lipid profile levels and amelioration of histological alterations in rats with diabetes. The administration of allantoin to diabetic rats resulted in a notable decrease in Malondialdehyde (MDA) levels, accompanied by an increase in the activity of antioxidant enzymes in the serum, liver, and kidney. The findings of oral glucose tolerance and hyperinsulinemic-euglycemic clamp tests demonstrated a significant rise in insulin resistance following the administration of allantoin. The upregulation of IRS-2/PI3K/p-Akt/GLUT expression by allantoin suggests a mechanistic relationship between the PI3K/Akt signaling pathway and the antihyperglycemic activity of allantoin. Furthermore, it resulted in a reduction in the levels of TGF-β1/p38MAPK/Caspase-3 expression in the aforementioned rat tissues affected by diabetes. Conclusions This study implies that allantoin treats type 2 diabetes by activating PI3K. Additionally, it reduces liver, kidney, and pancreatic apoptosis and inflammation-induced insulin resistance.re.
Collapse
Affiliation(s)
- Yao Zhao
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830017, Xinjiang, China
| | - Ming Qiao
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830000, Xinjiang, China
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi 830011, Xinjiang, China
| | - Xiaomei Wang
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830017, Xinjiang, China
| | - Xinjie Luo
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830017, Xinjiang, China
| | - Jianhua Yang
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830000, Xinjiang, China
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi 830011, Xinjiang, China
| | - Junping Hu
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830017, Xinjiang, China
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi 830011, Xinjiang, China
| |
Collapse
|
12
|
McKenna CF, Stierwalt HD, Zemski Berry KA, Ehrlicher SE, Robinson MM, Zarini S, Kahn DE, Snell-Bergeon JK, Perreault L, Bergman BC, Newsom SA. Intramuscular diacylglycerol accumulates with acute hyperinsulinemia in insulin-resistant phenotypes. Am J Physiol Endocrinol Metab 2024; 327:E183-E193. [PMID: 38895980 PMCID: PMC11427097 DOI: 10.1152/ajpendo.00368.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024]
Abstract
Elevated skeletal muscle diacylglycerols (DAGs) and ceramides can impair insulin signaling, and acylcarnitines (acylCNs) reflect impaired mitochondrial fatty acid oxidation, thus, the intramuscular lipid profile is indicative of insulin resistance. Acute (i.e., postprandial) hyperinsulinemia has been shown to elevate lipid concentrations in healthy muscle and is an independent risk factor for type 2 diabetes (T2D). However, it is unclear how the relationship between acute hyperinsulinemia and the muscle lipidome interacts across metabolic phenotypes, thus contributing to or exacerbating insulin resistance. We therefore investigated the impact of acute hyperinsulinemia on the skeletal muscle lipid profile to help characterize the physiological basis in which hyperinsulinemia elevates T2D risk. In a cross-sectional comparison, endurance athletes (n = 12), sedentary lean adults (n = 12), and individuals with obesity (n = 13) and T2D (n = 7) underwent a hyperinsulinemic-euglycemic clamp with muscle biopsies. Although there were no significant differences in total 1,2-DAG fluctuations, there was a 2% decrease in athletes versus a 53% increase in T2D during acute hyperinsulinemia (P = 0.087). Moreover, C18 1,2-DAG species increased during the clamp with T2D only, which negatively correlated with insulin sensitivity (P < 0.050). Basal muscle C18:0 total ceramides were elevated with T2D (P = 0.029), but not altered by clamp. Acylcarnitines were universally lowered during hyperinsulinemia, with more robust reductions of 80% in athletes compared with only 46% with T2D (albeit not statistically significant, main effect of group, P = 0.624). Similar fluctuations with acute hyperinsulinemia increasing 1,2 DAGs in insulin-resistant phenotypes and universally lowering acylcarnitines were observed in male mice. In conclusion, acute hyperinsulinemia elevates muscle 1,2-DAG levels with insulin-resistant phenotypes. This suggests a possible dysregulation of intramuscular lipid metabolism in the fed state in individuals with low insulin sensitivity, which may exacerbate insulin resistance.NEW & NOTEWORTHY Postprandial hyperinsulinemia is a risk factor for type 2 diabetes and may increase muscle lipids. However, it is unclear how the relationship between acute hyperinsulinemia and the muscle lipidome interacts across metabolic phenotypes, thus contributing to insulin resistance. We observed that acute hyperinsulinemia elevates muscle 1,2-DAGs in insulin-resistant phenotypes, whereas ceramides were unaltered. Insulin-mediated acylcarnitine reductions are also hindered with high-fat feeding. The postprandial period may exacerbate insulin resistance in metabolically unhealthy phenotypes.
Collapse
Affiliation(s)
- Colleen F McKenna
- Division of Endocrinology, Metabolism and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Harrison D Stierwalt
- School of Exercise, Sport, and Health Sciences, College of Health, Oregon State University, Corvallis, Oregon, United States
| | - Karin A Zemski Berry
- Division of Endocrinology, Metabolism and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Sarah E Ehrlicher
- School of Exercise, Sport, and Health Sciences, College of Health, Oregon State University, Corvallis, Oregon, United States
| | - Matthew M Robinson
- School of Exercise, Sport, and Health Sciences, College of Health, Oregon State University, Corvallis, Oregon, United States
| | - Simona Zarini
- Division of Endocrinology, Metabolism and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Darcy E Kahn
- Division of Endocrinology, Metabolism and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Janet K Snell-Bergeon
- Division of Endocrinology, Metabolism and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Leigh Perreault
- Division of Endocrinology, Metabolism and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Bryan C Bergman
- Division of Endocrinology, Metabolism and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Sean A Newsom
- School of Exercise, Sport, and Health Sciences, College of Health, Oregon State University, Corvallis, Oregon, United States
| |
Collapse
|
13
|
Vieira-Lara MA, Bakker BM. The paradox of fatty-acid β-oxidation in muscle insulin resistance: Metabolic control and muscle heterogeneity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167172. [PMID: 38631409 DOI: 10.1016/j.bbadis.2024.167172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/18/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
The skeletal muscle is a metabolically heterogeneous tissue that plays a key role in maintaining whole-body glucose homeostasis. It is well known that muscle insulin resistance (IR) precedes the development of type 2 diabetes. There is a consensus that the accumulation of specific lipid species in the tissue can drive IR. However, the role of the mitochondrial fatty-acid β-oxidation in IR and, consequently, in the control of glucose uptake remains paradoxical: interventions that either inhibit or activate fatty-acid β-oxidation have been shown to prevent IR. We here discuss the current theories and evidence for the interplay between β-oxidation and glucose uptake in IR. To address the underlying intricacies, we (1) dive into the control of glucose uptake fluxes into muscle tissues using the framework of Metabolic Control Analysis, and (2) disentangle concepts of flux and catalytic capacities taking into account skeletal muscle heterogeneity. Finally, we speculate about hitherto unexplored mechanisms that could bring contrasting evidence together. Elucidating how β-oxidation is connected to muscle IR and the underlying role of muscle heterogeneity enhances disease understanding and paves the way for new treatments for type 2 diabetes.
Collapse
Affiliation(s)
- Marcel A Vieira-Lara
- Laboratory of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Barbara M Bakker
- Laboratory of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
14
|
Batterson PM, McGowan EM, Borowik AK, Kinter MT, Miller BF, Newsom SA, Robinson MM. High-fat diet increases electron transfer flavoprotein synthesis and lipid respiration in skeletal muscle during exercise training in female mice. Physiol Rep 2023; 11:e15840. [PMID: 37857571 PMCID: PMC10587055 DOI: 10.14814/phy2.15840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/21/2023] Open
Abstract
High-fat diet (HFD) and exercise remodel skeletal muscle mitochondria. The electron transfer flavoproteins (ETF) transfer reducing equivalents from β-oxidation into the electron transfer system. Exercise may stimulate the synthesis of ETF proteins to increase lipid respiration. We determined mitochondrial remodeling for lipid respiration through ETF in the context of higher mitochondrial abundance/capacity seen in female mice. We hypothesized HFD would be a greater stimulus than exercise to remodel ETF and lipid pathways through increased protein synthesis alongside increased lipid respiration. Female C57BL/6J mice (n = 15 per group) consumed HFD or low-fat diet (LFD) for 4 weeks then remained sedentary (SED) or completed 8 weeks of treadmill training (EX). We determined mitochondrial lipid respiration, RNA abundance, individual protein synthesis, and abundance for ETFα, ETFβ, and ETF dehydrogenase (ETFDH). HFD increased absolute and relative lipid respiration (p = 0.018 and p = 0.034) and RNA abundance for ETFα (p = 0.026), ETFβ (p = 0.003), and ETFDH (p = 0.0003). HFD increased synthesis for ETFα and ETFDH (p = 0.0007 and p = 0.002). EX increased synthesis of ETFβ and ETFDH (p = 0.008 and p = 0.006). Higher synthesis rates of ETF were not always reflected in greater protein abundance. Greater synthesis of ETF during HFD indicates mitochondrial remodeling which may contribute higher mitochondrial lipid respiration through enhanced ETF function.
Collapse
Affiliation(s)
- Philip M. Batterson
- School of Biological and Population Health SciencesOregon State UniversityCorvallisOregonUSA
| | - Erin M. McGowan
- School of Biological and Population Health SciencesOregon State UniversityCorvallisOregonUSA
| | - Agnieszka K. Borowik
- Aging and Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Michael T. Kinter
- Aging and Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Benjamin F. Miller
- Aging and Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
- Oklahoma City VAOklahoma CityOklahomaUSA
| | - Sean A. Newsom
- School of Biological and Population Health SciencesOregon State UniversityCorvallisOregonUSA
| | - Matthew M. Robinson
- School of Biological and Population Health SciencesOregon State UniversityCorvallisOregonUSA
| |
Collapse
|
15
|
Ruocco C, Malavazos AE, Ragni M, Carruba MO, Valerio A, Iacobellis G, Nisoli E. Amino acids contribute to adaptive thermogenesis. New insights into the mechanisms of action of recent drugs for metabolic disorders are emerging. Pharmacol Res 2023; 195:106892. [PMID: 37619907 DOI: 10.1016/j.phrs.2023.106892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/28/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
Adaptive thermogenesis is the heat production by muscle contractions (shivering thermogenesis) or brown adipose tissue (BAT) and beige fat (non-shivering thermogenesis) in response to external stimuli, including cold exposure. BAT and beige fat communicate with peripheral organs and the brain through a variegate secretory and absorption processes - controlling adipokines, microRNAs, extracellular vesicles, and metabolites - and have received much attention as potential therapeutic targets for managing obesity-related disorders. The sympathetic nervous system and norepinephrine-releasing adipose tissue macrophages (ATM) activate uncoupling protein 1 (UCP1), expressed explicitly in brown and beige adipocytes, dissolving the electrochemical gradient and uncoupling tricarboxylic acid cycle and the electron transport chain from ATP production. Mounting evidence has attracted attention to the multiple effects of dietary and endogenously synthesised amino acids in BAT thermogenesis and metabolic phenotype in animals and humans. However, the mechanisms implicated in these processes have yet to be conclusively characterized. In the present review article, we aim to define the principal investigation areas in this context, including intestinal microbiota constitution, adipose autophagy modulation, and secretome and metabolic fluxes control, which lead to increased brown/beige thermogenesis. Finally, also based on our recent epicardial adipose tissue results, we summarise the evidence supporting the notion that the new dual and triple agonists of glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), and glucagon (GCG) receptor - with never before seen weight loss and insulin-sensitizing efficacy - promote thermogenic-like amino acid profiles in BAT with robust heat production and likely trigger sympathetic activation and adaptive thermogenesis by controlling amino acid metabolism and ATM expansion in BAT and beige fat.
Collapse
Affiliation(s)
- Chiara Ruocco
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy
| | - Alexis Elias Malavazos
- Endocrinology Unit, Clinical Nutrition and Cardiovascular Prevention Service, IRCCS Policlinico San Donato, Piazza Edmondo Malan, 2, San Donato Milanese, 20097 Milan, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, via della Commenda, 10, 20122 Milan, Italy
| | - Maurizio Ragni
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy
| | - Michele O Carruba
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy
| | - Alessandra Valerio
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa, 11, 25123 Brescia, Italy
| | - Gianluca Iacobellis
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami, 1400 NW 12th Ave, Miami, FL, USA
| | - Enzo Nisoli
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy.
| |
Collapse
|
16
|
Akingbesote ND, Leitner BP, Jovin DG, Desrouleaux R, Owusu D, Zhu W, Li Z, Pollak MN, Perry RJ. Gene and protein expression and metabolic flux analysis reveals metabolic scaling in liver ex vivo and in vivo. eLife 2023; 12:e78335. [PMID: 37219930 PMCID: PMC10205083 DOI: 10.7554/elife.78335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/08/2023] [Indexed: 05/24/2023] Open
Abstract
Metabolic scaling, the inverse correlation of metabolic rates to body mass, has been appreciated for more than 80 years. Studies of metabolic scaling have largely been restricted to mathematical modeling of caloric intake and oxygen consumption, and mostly rely on computational modeling. The possibility that other metabolic processes scale with body size has not been comprehensively studied. To address this gap in knowledge, we employed a systems approach including transcriptomics, proteomics, and measurement of in vitro and in vivo metabolic fluxes. Gene expression in livers of five species spanning a 30,000-fold range in mass revealed differential expression according to body mass of genes related to cytosolic and mitochondrial metabolic processes, and to detoxication of oxidative damage. To determine whether flux through key metabolic pathways is ordered inversely to body size, we applied stable isotope tracer methodology to study multiple cellular compartments, tissues, and species. Comparing C57BL/6 J mice with Sprague-Dawley rats, we demonstrate that while ordering of metabolic fluxes is not observed in in vitro cell-autonomous settings, it is present in liver slices and in vivo. Together, these data reveal that metabolic scaling extends beyond oxygen consumption to other aspects of metabolism, and is regulated at the level of gene and protein expression, enzyme activity, and substrate supply.
Collapse
Affiliation(s)
- Ngozi D Akingbesote
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Internal Medicine – Endocrinology, Yale UniversityNew HavenUnited States
| | - Brooks P Leitner
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Internal Medicine – Endocrinology, Yale UniversityNew HavenUnited States
| | - Daniel G Jovin
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Internal Medicine – Endocrinology, Yale UniversityNew HavenUnited States
| | - Reina Desrouleaux
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Comparative Medicine, Yale UniversityNew HavenUnited States
| | - Dennis Owusu
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Internal Medicine – Endocrinology, Yale UniversityNew HavenUnited States
| | - Wanling Zhu
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Internal Medicine – Endocrinology, Yale UniversityNew HavenUnited States
| | - Zongyu Li
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Internal Medicine – Endocrinology, Yale UniversityNew HavenUnited States
| | - Michael N Pollak
- Lady Davis Institute for Medical Research, Jewish General HospitalMontrealCanada
- Department of Oncology, McGill UniversityMontrealCanada
| | - Rachel J Perry
- Department of Cellular & Molecular Physiology, Yale UniversityNew HavenUnited States
- Department of Internal Medicine – Endocrinology, Yale UniversityNew HavenUnited States
| |
Collapse
|
17
|
Gaspar RC, Lyu K, Hubbard BT, Leitner BP, Luukkonen PK, Hirabara SM, Sakuma I, Nasiri A, Zhang D, Kahn M, Cline GW, Pauli JR, Perry RJ, Petersen KF, Shulman GI. Distinct subcellular localisation of intramyocellular lipids and reduced PKCε/PKCθ activity preserve muscle insulin sensitivity in exercise-trained mice. Diabetologia 2023; 66:567-578. [PMID: 36456864 PMCID: PMC11194860 DOI: 10.1007/s00125-022-05838-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 09/30/2022] [Indexed: 12/03/2022]
Abstract
AIMS/HYPOTHESIS Athletes exhibit increased muscle insulin sensitivity, despite increased intramuscular triacylglycerol content. This phenomenon has been coined the 'athlete's paradox' and is poorly understood. Recent findings suggest that the subcellular distribution of sn-1,2-diacylglycerols (DAGs) in the plasma membrane leading to activation of novel protein kinase Cs (PKCs) is a crucial pathway to inducing insulin resistance. Here, we hypothesised that regular aerobic exercise would preserve muscle insulin sensitivity by preventing increases in plasma membrane sn-1,2-DAGs and activation of PKCε and PKCθ despite promoting increases in muscle triacylglycerol content. METHODS C57BL/6J mice were allocated to three groups (regular chow feeding [RC]; high-fat diet feeding [HFD]; RC feeding and running wheel exercise [RC-EXE]). We used a novel LC-MS/MS/cellular fractionation method to assess DAG stereoisomers in five subcellular compartments (plasma membrane [PM], endoplasmic reticulum, mitochondria, lipid droplets and cytosol) in the skeletal muscle. RESULTS We found that the HFD group had a greater content of sn-DAGs and ceramides in multiple subcellular compartments compared with the RC mice, which was associated with an increase in PKCε and PKCθ translocation. However, the RC-EXE mice showed, of particular note, a reduction in PM sn-1,2-DAG and ceramide content when compared with HFD mice. Consistent with the PM sn-1,2-DAG-novel PKC hypothesis, we observed an increase in phosphorylation of threonine1150 on the insulin receptor kinase (IRKT1150), and reductions in insulin-stimulated IRKY1162 phosphorylation and IRS-1-associated phosphoinositide 3-kinase activity in HFD compared with RC and RC-EXE mice, which are sites of PKCε and PKCθ action, respectively. CONCLUSIONS/INTERPRETATION These results demonstrate that lower PKCθ/PKCε activity and sn-1,2-DAG content, especially in the PM compartment, can explain the preserved muscle insulin sensitivity in RC-EXE mice.
Collapse
Affiliation(s)
- Rafael C Gaspar
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- School of Applied Science, University of Campinas, Limeira, SP, Brazil
| | - Kun Lyu
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Brandon T Hubbard
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Brooks P Leitner
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Panu K Luukkonen
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Sandro M Hirabara
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Institute of Physical Activity Science and Sports, Cruzeiro do Sul University, São Paulo, SP, Brazil
| | - Ikki Sakuma
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Ali Nasiri
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Dongyan Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Mario Kahn
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Gary W Cline
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Rachel J Perry
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Kitt F Petersen
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
18
|
Batterson PM, McGowan EM, Stierwalt HD, Ehrlicher SE, Newsom SA, Robinson MM. Two weeks of high-intensity interval training increases skeletal muscle mitochondrial respiration via complex-specific remodeling in sedentary humans. J Appl Physiol (1985) 2023; 134:339-355. [PMID: 36603044 DOI: 10.1152/japplphysiol.00467.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Aerobic training remodels the quantity and quality (function per unit) of skeletal muscle mitochondria to promote substrate oxidation, however, there remain key gaps in understanding the underlying mechanisms during initial training adaptations. We used short-term high-intensity interval training (HIIT) to determine changes to mitochondrial respiration and regulatory pathways that occur early in remodeling. Fifteen normal-weight sedentary adults started seven sessions of HIIT over 14 days and 14 participants completed the intervention. We collected vastus lateralis biopsies before and 48 h after HIIT to determine mitochondrial respiration, RNA sequencing, and Western blotting for proteins of mitochondrial respiration and degradation via autophagy. HIIT increased respiration per mitochondrial protein for lipid (+23% P = 0.020), complex I (+18%, P = 0.0015), complex I + II (+14%, P < 0.0001), and complex II (+24% P < 0.0001). Transcripts that increased with HIIT identified several gene sets of mitochondrial respiration, particularly for complex I, whereas transcripts that decreased identified pathways of DNA and chromatin remodeling. HIIT lowered protein abundance of autophagy markers for p62 (-19%, P = 0.012) and LC3 II/I (-20%, P = 0.004) in whole tissue lysates but not isolated mitochondria. Meal tolerance testing revealed HIIT increased the change in whole body respiratory exchange ratio and lowered cumulative plasma insulin concentrations. Gene transcripts and respiratory function indicate remodeling of mitochondria within 2 wk of HIIT. Overall changes are consistent with increased protein quality driving rapid improvements in substrate oxidation.NEW & NOTEWORTHY Aerobic training stimulates mitochondrial metabolism in skeletal muscle that is linked to improvements to whole body fuel metabolism. The mechanisms driving changes to the quantity and quality (function per unit) of mitochondria are less known. We used seven sessions of high-intensity interval training (HIIT) to determine functional changes and mechanisms of mitochondrial remodeling in skeletal muscle. HIIT increased mitochondrial respiration per mass for fatty acids, complex I, and complex II substrates. HIIT-induced remodeling pathways including gene transcripts for mitochondrial respiration (via RNA sequencing of muscle tissue) and proteins related to complex I respiration. We conclude that an early feature of aerobic training is increased mitochondrial protein quality via improved respiration and induction of mitochondrial transcriptional patterns.
Collapse
Affiliation(s)
- Philip M Batterson
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| | - Erin M McGowan
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| | - Harrison D Stierwalt
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| | - Sarah E Ehrlicher
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| | - Sean A Newsom
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| | - Matthew M Robinson
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| |
Collapse
|
19
|
Villela Nogueira CA, Leite NC. Nonalcoholic Fatty Liver in the Pathogenesis of Diabetes. THE DIABETES TEXTBOOK 2023:261-270. [DOI: 10.1007/978-3-031-25519-9_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
20
|
Chong CR, Liu S, Imam H, Heresztyn T, Sallustio BC, Chirkov YY, Horowitz JD. Perhexiline Therapy in Patients with Type 2 Diabetes: Incremental Insulin Resistance despite Potentiation of Nitric Oxide Signaling. Biomedicines 2022; 10:2381. [PMID: 36289640 PMCID: PMC9598312 DOI: 10.3390/biomedicines10102381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/16/2022] [Accepted: 09/17/2022] [Indexed: 11/21/2022] Open
Abstract
Perhexiline (Px) inhibits carnitine palmitoyltransferase 1 (CPT1), which controls uptake of long chain fatty acids into mitochondria. However, occasional cases of hypoglycaemia have been reported in Px-treated patients, raising the possibility that Px may also increase sensitivity to insulin. Furthermore, Px increases anti-aggregatory responses to nitric oxide (NO), an effect which may theoretically parallel insulin sensitization. We therefore sought to examine these relationships in patients with stable Type 2 diabetes (T2D) and cardiovascular disease (n = 30). Px was initiated, and dosage was titrated, to reach the therapeutic range and thus prevent toxicity. Investigations were performed before and after 2 weeks, to examine changes in insulin sensitivity and, utilizing aggregometry in whole blood, platelet responsiveness to the anti-aggregatory effects of the NO donor sodium nitroprusside (SNP). Other parameters that affect may affect NO signalling were also evaluated. Px substantially potentiated inhibition of platelet aggregation by SNP (from 16.7 ± 3.0 to 27.3 ± 3.7%; p = 0.005). Px did not change fasting blood glucose concentrations but reduced insulin sensitivity (HOMA-IR score increased from median of 4.47 to 6.08; p = 0.028), and increased fasting plasma insulin concentrations (median 16.5 to 19.0 mU/L; p = 0.014). Increases in SNP responses tended (r = -0.30; p = 0.11) to be reciprocally related to increases in HOMA-IR, and increases in HOMA-IR were greater (p = 0.002) in patients without NO-sensitizing effects. No patient developed symptomatic hypoglycaemia, nor was there any other short-term toxicity of Px. Thus, in patients with stable T2D and cardiovascular disease, Px increases anti-aggregatory responsiveness to NO, but is not an insulin sensitizer, and does not induce hypoglycaemia. Absence of NO-sensitizing effect occurs in approximately 30% of Px-treated patients with T2D, and is associated with induction of insulin resistance in these patients.
Collapse
Affiliation(s)
- Cher-Rin Chong
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South 5011, Australia
| | - Saifei Liu
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South 5011, Australia
| | - Hasan Imam
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South 5011, Australia
| | - Tamila Heresztyn
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South 5011, Australia
| | - Benedetta C. Sallustio
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South 5011, Australia
- School of Medical Sciences, The University of Adelaide, Adelaide 5000, Australia
| | - Yuliy Y. Chirkov
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South 5011, Australia
| | - John D. Horowitz
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South 5011, Australia
| |
Collapse
|
21
|
Karppinen JE, Juppi HK, Hintikka J, Wiklund P, Haapala EA, Hyvärinen M, Tammelin TH, Aukee P, Kujala UM, Laukkanen J, Laakkonen EK. Associations of resting and peak fat oxidation with sex hormone profile and blood glucose control in middle-aged women. Nutr Metab Cardiovasc Dis 2022; 32:2157-2167. [PMID: 35752543 DOI: 10.1016/j.numecd.2022.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND AIMS Menopause may reduce fat oxidation. We investigated whether sex hormone profile explains resting fat oxidation (RFO) or peak fat oxidation (PFO) during incremental cycling in middle-aged women. Secondarily, we studied associations of RFO and PFO with glucose regulation. METHOD AND RESULTS We measured RFO and PFO of 42 women (age 52-58 years) with indirect calorimetry. Seven participants were pre- or perimenopausal, 26 were postmenopausal, and nine were postmenopausal hormone therapy users. Serum estradiol (E2), follicle-stimulating hormone, progesterone, and testosterone levels were quantified with immunoassays. Insulin sensitivity (Matsuda index) and glucose tolerance (area under the curve) were determined by glucose tolerance testing. Body composition was assessed with dual-energy X-ray absorptiometry; physical activity with self-report and accelerometry; and diet, with food diaries. Menopausal status or sex hormone levels were not associated with the fat oxidation outcomes. RFO determinants were fat mass (β = 0.44, P = 0.006) and preceding energy intake (β = -0.40, P = 0.019). Cardiorespiratory fitness (β = 0.59, P = 0.002), lean mass (β = 0.49, P = 0.002) and physical activity (self-reported β = 0.37, P = 0.020; accelerometer-measured β = 0.35, P = 0.024) explained PFO. RFO and PFO were not related to insulin sensitivity. Higher RFO was associated with poorer glucose tolerance (β = 0.52, P = 0.002). CONCLUSION Among studied middle-aged women, sex hormone profile did not explain RFO or PFO, and higher fat oxidation capacity did not indicate better glucose control.
Collapse
Affiliation(s)
- Jari E Karppinen
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.
| | - Hanna-Kaarina Juppi
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Jukka Hintikka
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Petri Wiklund
- Exercise Translational Medicine Center, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China; Huawei Helsinki R&D Center, Huawei Technologies Ltd, Helsinki, Finland
| | - Eero A Haapala
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland; Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Matti Hyvärinen
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | | | - Pauliina Aukee
- Department of Obstetrics and Gynecology, Central Finland Health Care District, Jyväskylä, Finland
| | - Urho M Kujala
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Jari Laukkanen
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland; Department of Internal Medicine, Central Finland Healthcare District, Jyväskylä, Finland; Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Eija K Laakkonen
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
22
|
Mitochondria-Mediated Cardiovascular Benefits of Sodium-Glucose Co-Transporter 2 Inhibitors. Int J Mol Sci 2022; 23:ijms23105371. [PMID: 35628174 PMCID: PMC9140946 DOI: 10.3390/ijms23105371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 11/23/2022] Open
Abstract
Several recent cardiovascular trials of SGLT 2 (sodium-glucose cotransporter 2) inhibitors revealed that they could reduce adverse cardiovascular events in patients with T2DM (type 2 diabetes mellitus). However, the exact molecular mechanism underlying the beneficial effects that SGLT2 inhibitors have on the cardiovascular system is still unknown. In this review, we focus on the molecular mechanisms of the mitochondria-mediated beneficial effects of SGLT2 inhibitors on the cardiovascular system. The application of SGLT2 inhibitors ameliorates mitochondrial dysfunction, dynamics, bioenergetics, and ion homeostasis and reduces the production of mitochondrial reactive oxygen species, which results in cardioprotective effects. Herein, we present a comprehensive overview of the impact of SGLT2 inhibitors on mitochondria and highlight the potential application of these medications to treat both T2DM and cardiovascular diseases.
Collapse
|
23
|
Palmer BF, Clegg DJ. Metabolic Flexibility and Its Impact on Health Outcomes. Mayo Clin Proc 2022; 97:761-776. [PMID: 35287953 DOI: 10.1016/j.mayocp.2022.01.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 01/04/2022] [Accepted: 01/12/2022] [Indexed: 02/06/2023]
Abstract
A metabolically flexible state exists when there is a rapid switch between glucose and fatty acids during the transition between the fed and fasting state. This flexibility in fuel choice serves to prevent hyperglycemia following a meal and simultaneously ensures an adequate amount of blood glucose is available for delivery to the brain and exclusively glycolytic tissues during fasting. The modern era is characterized by chronic overnutrition in which a mixture of fuels is delivered to the mitochondria in an unabated manner thereby uncoupling the feast and famine situation. The continuous influx of fuel leads to accumulation of reducing equivalents in the mitochondria and an increase in the mitochondrial membrane potential. These changes create a microenvironment fostering the generation of reactive oxygen species and other metabolites leading to deleterious protein modification, cell injury, and ultimately clinical disease. Insulin resistance may also play a primary role in this deleterious effect. The imbalance between mitochondrial energy delivery and use is made worse with a sedentary lifestyle. Maneuvers that restore energy balance across the mitochondria activate pathways that remove or repair damaged molecules and restore the plasticity characteristic of normal energy metabolism. Readily available strategies to maintain energy balance across the mitochondria include exercise, various forms of caloric restriction, administration of sodium-glucose cotransporter-2 inhibitors, cold exposure, and hypobaric hypoxia.
Collapse
Affiliation(s)
- Biff F Palmer
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | | |
Collapse
|
24
|
DeVito LM, Dennis EA, Kahn BB, Shulman GI, Witztum JL, Sadhu S, Nickels J, Spite M, Smyth S, Spiegel S. Bioactive lipids and metabolic syndrome-a symposium report. Ann N Y Acad Sci 2022; 1511:87-106. [PMID: 35218041 DOI: 10.1111/nyas.14752] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 11/27/2022]
Abstract
Recent research has shed light on the cellular and molecular functions of bioactive lipids that go far beyond what was known about their role as dietary lipids. Bioactive lipids regulate inflammation and its resolution as signaling molecules. Genetic studies have identified key factors that can increase the risk of cardiovascular diseases and metabolic syndrome through their effects on lipogenesis. Lipid scientists have explored how these signaling pathways affect lipid metabolism in the liver, adipose tissue, and macrophages by utilizing a variety of techniques in both humans and animal models, including novel lipidomics approaches and molecular dynamics models. Dissecting out these lipid pathways can help identify mechanisms that can be targeted to prevent or treat cardiometabolic conditions. Continued investigation of the multitude of functions mediated by bioactive lipids may reveal additional components of these pathways that can provide a greater understanding of metabolic homeostasis.
Collapse
Affiliation(s)
| | | | - Barbara B Kahn
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | | | | | | | - Joseph Nickels
- Genesis Biotechnology Group, Hamilton Township, New Jersey
| | - Matthew Spite
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Susan Smyth
- University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Sarah Spiegel
- Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
25
|
Affiliation(s)
- Aaron M Cypess
- From the Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
26
|
Experimental models of lipid overload and their relevance in understanding skeletal muscle insulin resistance and pathological changes in mitochondrial oxidative capacity. Biochimie 2021; 196:182-193. [PMID: 34563603 DOI: 10.1016/j.biochi.2021.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/30/2021] [Accepted: 09/20/2021] [Indexed: 02/08/2023]
Abstract
It remains essential to decipher some of the pathological mechanisms that link obesity with deteriorating human health. Insulin resistance, due to enhanced free fatty acid substrate delivery, results in disrupted glucose homeostasis and altered mitochondrial oxidative capacity, which is a characteristic feature of an obese state. In fact, as a major site for regulating glucose homeostasis and energy production in response to insulin, the skeletal muscle has become an interesting target tissue to understand the impact of lipid overload on the development of insulin resistance and impaired mitochondrial respiratory function. In addition to systematically retrieving the discussed data, the current review brings an essential perspective in understanding the relevance of experimental models of lipid overload such as high fat diets in understanding the pathological link between insulin resistance and pathological changes in mitochondrial oxidative capacity. Importantly, inclusion of evidence from transgenic model highlights some of the unique molecular targets that are implicated in the development of insulin resistance and inefficient mitochondrial respiration processes within an obese state. Importantly, saturation with lipid products such as ceramides and diacylglycerols, especially within the skeletal muscle, appears to be instrumental in paving the path leading to worsening of metabolic complications. These metabolic consequences mostly interfere with the efficiency of the mitochondrial electron transport chain, leading to overproduction of toxic reactive oxygen species. Therefore, therapeutic agents that reverse the effects of lipid overload by improving insulin sensitivity and mitochondrial oxidative capacity are crucial for the management or even treatment of metabolic diseases.
Collapse
|
27
|
Bednarski TK, Rahim M, Young JD. In vivo 2H/ 13C flux analysis in metabolism research. Curr Opin Biotechnol 2021; 71:1-8. [PMID: 34048994 DOI: 10.1016/j.copbio.2021.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/29/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022]
Abstract
Identifying the factors and mechanisms that regulate metabolism under normal and diseased states requires methods to quantify metabolic fluxes of live tissues within their physiological milieu. A number of recent developments have expanded the reach and depth of isotope-based in vivo flux analysis, which have in turn challenged existing dogmas in metabolism research. First, minimally invasive techniques of intravenous isotope infusion and sampling have advanced in vivo metabolic tracer studies in animal models and human subjects. Second, recent breakthroughs in analytical instrumentation have expanded the scope of isotope labeling measurements and reduced sample volume requirements. Third, innovative modeling approaches and publicly available software tools have facilitated rigorous analysis of sophisticated experimental designs involving multiple tracers and expansive metabolomics datasets. These developments have enabled comprehensive in vivo quantification of metabolic fluxes in specific tissues and have set the stage for integrated multi-tissue flux assays.
Collapse
Affiliation(s)
- Tomasz K Bednarski
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Mohsin Rahim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
28
|
Loomba R, Friedman SL, Shulman GI. Mechanisms and disease consequences of nonalcoholic fatty liver disease. Cell 2021; 184:2537-2564. [PMID: 33989548 PMCID: PMC12168897 DOI: 10.1016/j.cell.2021.04.015] [Citation(s) in RCA: 1136] [Impact Index Per Article: 284.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/21/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the leading chronic liver disease worldwide. Its more advanced subtype, nonalcoholic steatohepatitis (NASH), connotes progressive liver injury that can lead to cirrhosis and hepatocellular carcinoma. Here we provide an in-depth discussion of the underlying pathogenetic mechanisms that lead to progressive liver injury, including the metabolic origins of NAFLD, the effect of NAFLD on hepatic glucose and lipid metabolism, bile acid toxicity, macrophage dysfunction, and hepatic stellate cell activation, and consider the role of genetic, epigenetic, and environmental factors that promote fibrosis progression and risk of hepatocellular carcinoma in NASH.
Collapse
Affiliation(s)
- Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology, Department of Medicine, University of California at San Diego, La Jolla, CA 92093, USA.
| | - Scott L Friedman
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale Diabetes Research Center, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
29
|
Pillon NJ, Loos RJF, Marshall SM, Zierath JR. Metabolic consequences of obesity and type 2 diabetes: Balancing genes and environment for personalized care. Cell 2021; 184:1530-1544. [PMID: 33675692 PMCID: PMC9191863 DOI: 10.1016/j.cell.2021.02.012] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/25/2020] [Accepted: 02/03/2021] [Indexed: 02/07/2023]
Abstract
The prevalence of type 2 diabetes and obesity has risen dramatically for decades and is expected to rise further, secondary to the growing aging, sedentary population. The strain on global health care is projected to be colossal. This review explores the latest work and emerging ideas related to genetic and environmental factors influencing metabolism. Translational research and clinical applications, including the impact of the COVID-19 pandemic, are highlighted. Looking forward, strategies to personalize all aspects of prevention, management and care are necessary to improve health outcomes and reduce the impact of these metabolic diseases.
Collapse
Affiliation(s)
- Nicolas J Pillon
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sally M Marshall
- Diabetes Research Group, Translational and Clinical Research Institute, Faculty of Clinical Medical Sciences, Newcastle University, 4(th) Floor William Leech Building, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
30
|
Lyu K, Zhang D, Song J, Li X, Perry RJ, Samuel VT, Shulman GI. Short-term overnutrition induces white adipose tissue insulin resistance through sn-1,2-diacylglycerol/PKCε/insulin receptor Thr1160 phosphorylation. JCI Insight 2021; 6:139946. [PMID: 33411692 PMCID: PMC7934919 DOI: 10.1172/jci.insight.139946] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 12/29/2020] [Indexed: 12/17/2022] Open
Abstract
White adipose tissue (WAT) insulin action has critical anabolic function and is dysregulated in overnutrition. However, the mechanism of short-term high-fat diet-induced (HFD-induced) WAT insulin resistance (IR) is poorly understood. Based on recent evidences, we hypothesize that a short-term HFD causes WAT IR through plasma membrane (PM) sn-1,2-diacylglycerol (sn-1,2-DAG) accumulation, which promotes protein kinase C-ε (PKCε) activation to impair insulin signaling by phosphorylating insulin receptor (Insr) Thr1160. To test this hypothesis, we assessed WAT insulin action in 7-day HFD-fed versus regular chow diet-fed rats during a hyperinsulinemic-euglycemic clamp. HFD feeding caused WAT IR, reflected by impaired insulin-mediated WAT glucose uptake and lipolysis suppression. These changes were specifically associated with PM sn-1,2-DAG accumulation, higher PKCε activation, and impaired insulin-stimulated Insr Tyr1162 phosphorylation. In order to examine the role of Insr Thr1160 phosphorylation in mediating lipid-induced WAT IR, we examined these same parameters in InsrT1150A mice (mouse homolog for human Thr1160) and found that HFD feeding induced WAT IR in WT control mice but not in InsrT1150A mice. Taken together, these data demonstrate the importance of the PM sn-1,2-DAG/PKCε/Insr Thr1160 phosphorylation pathway in mediating lipid-induced WAT IR and represent a potential therapeutic target to improve WAT insulin sensitivity.
Collapse
Affiliation(s)
- Kun Lyu
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Dongyan Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Joongyu Song
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Xiruo Li
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Rachel J Perry
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Varman T Samuel
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA.,VA Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
31
|
Yin L, Luo M, Wang R, Ye J, Wang X. Mitochondria in Sex Hormone-Induced Disorder of Energy Metabolism in Males and Females. Front Endocrinol (Lausanne) 2021; 12:749451. [PMID: 34987473 PMCID: PMC8721233 DOI: 10.3389/fendo.2021.749451] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 11/24/2021] [Indexed: 01/01/2023] Open
Abstract
Androgens have a complex role in the regulation of insulin sensitivity in the pathogenesis of type 2 diabetes. In male subjects, a reduction in androgens increases the risk for insulin resistance, which is improved by androgen injections. However, in female subjects with polycystic ovary syndrome (PCOS), androgen excess becomes a risk factor for insulin resistance. The exact mechanism underlying the complex activities of androgens remains unknown. In this review, a hormone synergy-based view is proposed for understanding this complexity. Mitochondrial overactivation by substrate influx is a mechanism of insulin resistance in obesity. This concept may apply to the androgen-induced insulin resistance in PCOS. Androgens and estrogens both exhibit activities in the induction of mitochondrial oxidative phosphorylation. The two hormones may synergize in mitochondria to induce overproduction of ATP. ATP surplus in the pancreatic β-cells and α-cells causes excess secretion of insulin and glucagon, respectively, leading to peripheral insulin resistance in the early phase of type 2 diabetes. In the skeletal muscle and liver, the ATP surplus contributes to insulin resistance through suppression of AMPK and activation of mTOR. Consistent ATP surplus leads to mitochondrial dysfunction as a consequence of mitophagy inhibition, which provides a potential mechanism for mitochondrial dysfunction in β-cells and brown adipocytes in PCOS. The hormone synergy-based view provides a basis for the overactivation and dysfunction of mitochondria in PCOS-associated type 2 diabetes. The molecular mechanism for the synergy is discussed in this review with a focus on transcriptional regulation. This view suggests a unifying mechanism for the distinct metabolic roles of androgens in the control of insulin action in men with hypogonadism and women with PCOS.
Collapse
Affiliation(s)
- Lijun Yin
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Man Luo
- Metabolism Research Center, Zhengzhou University Affiliated Zhengzhou Central Hospital, Zhengzhou, China
| | - Ru Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jianping Ye
- Metabolism Research Center, Zhengzhou University Affiliated Zhengzhou Central Hospital, Zhengzhou, China
- Center for Advanced Medicine, College of Medicine, Zhengzhou University, Zhengzhou, China
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- *Correspondence: Jianping Ye, ; Xiaohui Wang,
| | - Xiaohui Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- *Correspondence: Jianping Ye, ; Xiaohui Wang,
| |
Collapse
|
32
|
Mechanisms by which adiponectin reverses high fat diet-induced insulin resistance in mice. Proc Natl Acad Sci U S A 2020; 117:32584-32593. [PMID: 33293421 PMCID: PMC7768680 DOI: 10.1073/pnas.1922169117] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adiponectin has emerged as a potential therapy for type 2 diabetes mellitus, but the molecular mechanism by which adiponectin reverses insulin resistance remains unclear. Two weeks of globular adiponectin (gAcrp30) treatment reduced fasting plasma glucose, triglyceride (TAG), and insulin concentrations and reversed whole-body insulin resistance, which could be attributed to both improved insulin-mediated suppression of endogenous glucose production and increased insulin-stimulated glucose uptake in muscle and adipose tissues. These improvements in liver and muscle sensitivity were associated with ∼50% reductions in liver and muscle TAG and plasma membrane (PM)-associated diacylglycerol (DAG) content and occurred independent of reductions in total ceramide content. Reductions of PM DAG content in liver and skeletal muscle were associated with reduced PKCε translocation in liver and reduced PKCθ and PKCε translocation in skeletal muscle resulting in increased insulin-stimulated insulin receptor tyrosine1162 phosphorylation, IRS-1/IRS-2-associated PI3-kinase activity, and Akt-serine phosphorylation. Both gAcrp30 and full-length adiponectin (Acrp30) treatment increased eNOS/AMPK activation in muscle and muscle fatty acid oxidation. gAcrp30 and Acrp30 infusions also increased TAG uptake in epididymal white adipose tissue (eWAT), which could be attributed to increased lipoprotein lipase (LPL) activity. These data suggest that adiponectin and adiponectin-related molecules reverse lipid-induced liver and muscle insulin resistance by reducing ectopic lipid storage in these organs, resulting in decreased plasma membrane sn-1,2-DAG-induced nPKC activity and increased insulin signaling. Adiponectin mediates these effects by both promoting the storage of TAG in eWAT likely through stimulation of LPL as well as by stimulation of AMPK in muscle resulting in increased muscle fat oxidation.
Collapse
|
33
|
Kim IY, Park S, Kim Y, Chang Y, Choi CS, Suh SH, Wolfe RR. In Vivo and In Vitro Quantification of Glucose Kinetics: From Bedside to Bench. Endocrinol Metab (Seoul) 2020; 35:733-749. [PMID: 33397035 PMCID: PMC7803595 DOI: 10.3803/enm.2020.406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 11/19/2020] [Accepted: 11/27/2020] [Indexed: 12/16/2022] Open
Abstract
Like other substrates, plasma glucose is in a dynamic state of constant turnover (i.e., rates of glucose appearance [Ra glucose] into and disappearance [Rd glucose] from the plasma) while staying within a narrow range of normal concentrations, a physiological priority. Persistent imbalance of glucose turnover leads to elevations (i.e., hyperglycemia, Ra>Rd) or falls (i.e., hypoglycemia, Ra
Collapse
Affiliation(s)
- Il-Young Kim
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Seoul,
Korea
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Seoul,
Korea
| | - Sanghee Park
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Seoul,
Korea
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Seoul,
Korea
| | - Yeongmin Kim
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences & Technology (GAIHST), Gachon University, Incheon, Seoul,
Korea
| | - Yewon Chang
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences & Technology (GAIHST), Gachon University, Incheon, Seoul,
Korea
| | - Cheol Soo Choi
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Seoul,
Korea
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Seoul,
Korea
| | - Sang-Hoon Suh
- Department of Physical Education, Yonsei University, Seoul,
Korea
| | - Robert R. Wolfe
- Department of Geriatrics, the Center for Translational Research in Aging & Longevity, Donald W. Reynolds Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR,
USA
| |
Collapse
|