1
|
Peña de la Sancha P, Wieser BI, Schauer S, Reicher H, Sattler W, Breinbauer R, Schweiger M, Lechleitner M, Frank S, Zechner R, Kratky D, Espenshade PJ, Hoefler G, Vesely PW. Lipolysis-derived fatty acids are needed for homeostatic control of sterol element-binding protein-1c driven hepatic lipogenesis. Commun Biol 2025; 8:588. [PMID: 40205023 PMCID: PMC11982389 DOI: 10.1038/s42003-025-08002-1] [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: 11/15/2023] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
Sterol Regulatory Element-Binding Protein-1c (SREBP-1c) is translated as an inactive precursor (P-SREBP-1c) postprandially. Low levels of unsaturated fatty acids (uFAs) and high insulin promote its proteolytic activation, yielding N-SREBP-1c that drives fatty acid (FA) biosynthesis. During fasting, however, lipogenesis is low, and adipose tissue lipolysis supplies the organism with FAs. Adipose Triglyceride Lipase (ATGL) is the rate-limiting enzyme for adipose tissue lipolysis, and it preferentially releases uFAs. Therefore, we hypothesized that adipose ATGL-derived uFAs suppress P-SREBP-1c activation in the liver. In this study, we show that (I) N-SREBP-1c is transiently higher in livers of fasted and refed adipose specific Atgl knockout mice than in control livers. (II) This effect is reversed by injection of uFAs. (III) uFAs inhibit endoplasmic reticulum to Golgi-apparatus transport of SREBP Cleavage-Activating Protein (SCAP) in hepatocytes, which is essential for SREBP activation. Our findings demonstrate that adipose tissue ATGL derived uFAs attenuate P-SREBP-1c activation in the liver mainly after refeeding. We propose that this ATGL/SREBP-1c axis adds an additional layer of coordination between lipogenesis and lipolysis.
Collapse
Affiliation(s)
- Paola Peña de la Sancha
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Beatrix Irene Wieser
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Silvia Schauer
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Helga Reicher
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Wolfgang Sattler
- BioTechMed-Graz, Graz, Austria
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Rolf Breinbauer
- BioTechMed-Graz, Graz, Austria
- Institute of Organic Chemistry, Graz University of Technology, Graz, Austria
| | - Martina Schweiger
- BioTechMed-Graz, Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Margarete Lechleitner
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Saša Frank
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Rudolf Zechner
- BioTechMed-Graz, Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Dagmar Kratky
- BioTechMed-Graz, Graz, Austria
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Peter John Espenshade
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Gerald Hoefler
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Paul Willibald Vesely
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
- Otto Loewi Research Center, Lung Research Group, Graz, Austria.
| |
Collapse
|
2
|
Kumar GVN, Wang RS, Sharma AX, David NL, Amorim T, Sinden DS, Doshi NK, Wabitsch M, Gingras S, Ejaz A, Rubin JP, Maron BA, Fazeli PK, Steinhauser ML. Non-canonical lysosomal lipolysis drives mobilization of adipose tissue energy stores with fasting. Nat Commun 2025; 16:1330. [PMID: 39900947 PMCID: PMC11790841 DOI: 10.1038/s41467-025-56613-3] [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: 10/08/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Physiological adaptations to fasting enable humans to survive for prolonged periods without food and involve molecular pathways that may drive life-prolonging effects of dietary restriction in model organisms. Mobilization of fatty acids and glycerol from adipocyte lipid stores by canonical neutral lipases, including the rate limiting adipose triglyceride lipase (Pnpla2/ATGL), is critical to the adaptive fasting response. Here we discovered an alternative mechanism of lipolysis in adipocytes involving a lysosomal program. We functionally tested lysosomal lipolysis with pharmacological and genetic approaches in mice and in murine and human adipocyte and adipose tissue explant culture, establishing dependency on lysosomal acid lipase (LIPA/LAL) and the microphthalmia/transcription factor E (MiT/TFE) family. Our study establishes a model whereby the canonical pathway is critical for rapid lipolytic responses to adrenergic stimuli operative in the acute stage of fasting, while the alternative lysosomal pathway dominates with prolonged fasting.
Collapse
Affiliation(s)
- G V Naveen Kumar
- Aging Institute of UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rui-Sheng Wang
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ankit X Sharma
- Aging Institute of UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Natalie L David
- Aging Institute of UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tânia Amorim
- Aging Institute of UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Daniel S Sinden
- Aging Institute of UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nandini K Doshi
- Aging Institute of UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Martin Wabitsch
- University Medical Center Department of Pediatrics and Adolescent Medicine, Ulm, Germany
| | - Sebastien Gingras
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - J Peter Rubin
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Bradley A Maron
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- The University of Maryland-Institute for Health Computing, Bethesda, MD, USA
| | - Pouneh K Fazeli
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Matthew L Steinhauser
- Aging Institute of UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Moro C, Magnan C. Revisited guidelines for metabolic tolerance tests in mice. Lab Anim (NY) 2025; 54:16-23. [PMID: 39587363 PMCID: PMC11695259 DOI: 10.1038/s41684-024-01473-5] [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: 04/09/2024] [Accepted: 10/21/2024] [Indexed: 11/27/2024]
Abstract
Preclinical mouse models are extensively used in biomedical research to gain insight into disease mechanisms and to test new drug treatments. Glucose and insulin tolerance tests are simple experimental tests frequently used worldwide to assess glucose metabolism in mice. Various guidelines and methodological considerations have been published to help researchers standardize procedures and optimize research outcomes. Yet, there is still important experimental heterogeneity in the way these simple procedures are performed, with no real consensus on what the best practices are to achieve high-quality research and reproducible results. Here we critically examine several published guidelines and recent technical reports on how to perform these metabolic tests in laboratory mice and discuss the influence of various confounding factors on test results. We hope this work will help scientists establish more consensual guidelines for maximizing the relevance and clinical translation of studies using mouse models in metabolic research.
Collapse
Affiliation(s)
- Cedric Moro
- Institute of Metabolic and Cardiovascular Diseases, INSERM, Team MetaDiab, Paul Sabatier University, UMR1297, Toulouse, France.
| | - Christophe Magnan
- Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France
| |
Collapse
|
4
|
Ezzati‐Mobaser S, Yarahmadi S, Dadkhah Nikroo N, Maleki MH, Yousefi Z, Golpour P, Nourbakhsh M, Nourbakhsh M. Adipose triglyceride lipase gene expression in peripheral blood mononuclear cells of subjects with obesity and its association with insulin resistance, inflammation and lipid accumulation in liver. Obes Sci Pract 2024; 10:e716. [PMID: 38263987 PMCID: PMC10804332 DOI: 10.1002/osp4.716] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 01/25/2024] Open
Abstract
INTRODUCTION Adipose triglyceride lipase (ATGL) is a crucial enzyme responsible for the release of fatty acids from various tissues. The expression of ATGL is regulated by insulin and this enzyme is linked to Insulin resistance (IR). On the other hand, ATGL-mediated lipolysis is connected to macrophage function and thus, ATGL is involved in inflammation and the pathogenesis of lipid-related disorders. This study aimed to investigate the correlation between ATGL, obesity, Metabolic Syndrome (MetS), and inflammation. METHODS A total of 100 participants, including 50 individuals with obesity and 50 healthy participants, were recruited for this study and underwent comprehensive clinical evaluations. Blood samples were collected to measure plasma lipid profiles, glycemic indices, and liver function tests. Additionally, peripheral blood mononuclear cells (PBMCs) were isolated and used for the assessment of the gene expression of ATGL, using real-time PCR. Furthermore, PBMCs were cultured and exposed to lipopolysaccharides (LPS) with simultaneous ATGL inhibition, and the gene expression of inflammatory cytokines, along with the secretion of prostaglandin E2 (PGE2), were measured. RESULTS The gene expression of ATGL was significantly elevated in PBMCs obtained from participants with obesity and was particularly higher in those diagnosed with MetS. It exhibited a correlation with insulin levels and Homeostatic Model Assessment for IR (HOMA-IR), and it was associated with lipid accumulation in the liver. Stimulation with LPS increased ATGL expression in PBMCs, while inhibition of ATGL attenuated the inflammatory responses induced by LPS. CONCLUSIONS Obesity and MetS were associated with dysregulation of ATGL. ATGL might play a role in the upregulation of inflammatory cytokines and act as a significant contributor to the development of metabolic abnormalities related to obesity.
Collapse
Affiliation(s)
| | - Sahar Yarahmadi
- Department of BiochemistryFaculty of MedicineIran University of Medical SciencesTehranIran
| | - Nikta Dadkhah Nikroo
- Metabolic Disorders Research CenterEndocrinology and Metabolism Molecular‐Cellular Sciences InstituteTehran University of Medical SciencesTehranIran
| | - Mohammad Hasan Maleki
- Department of BiochemistrySchool of MedicineShiraz University of Medical SciencesShirazIran
| | - Zeynab Yousefi
- Department of Clinical BiochemistryFaculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Pegah Golpour
- Department of BiochemistryFaculty of MedicineShahid Sadoughi University of Medical SciencesTehranIran
| | - Mona Nourbakhsh
- Hazrat Aliasghar HospitalSchool of MedicineIran University of Medical SciencesTehranIran
| | - Mitra Nourbakhsh
- Finetech in Medicine Research CenterIran University of Medical SciencesTehranIran
- Department of BiochemistryFaculty of MedicineIran University of Medical SciencesTehranIran
| |
Collapse
|
5
|
Yu CHJ, Kienesberger PC, Pulinilkunnil T, Rupasinghe HPV. Effect of (poly)phenol-rich 'Daux Belan' apple supplementation on diet-induced obesity and glucose intolerance in C57BL/6NCrl mice. Sci Rep 2023; 13:17206. [PMID: 37821510 PMCID: PMC10567707 DOI: 10.1038/s41598-023-43687-6] [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: 06/12/2023] [Accepted: 09/27/2023] [Indexed: 10/13/2023] Open
Abstract
Obesity is a state of metabolic dysfunction that can lead to dyslipidemia and impaired glucose homeostasis. Apple polyphenols have been shown to ameliorate dyslipidemia/metabolic dysfunction in humans. The influence of apple (poly)phenols on energy metabolism in high-fat (HF) diet-induced obese mice remains controversial. This study examined the effect of dietary supplementation of (poly)phenol-rich 'Daux Belan' apple (DB; 6.2 mg gallic acid equivalence (GAE)/mouse/day; 0.15% (poly)phenol) in the form of freeze-dried powder on glucose and lipid metabolism in male HF-fed C57BL/6NCrl mice, in comparison to low-(poly)phenol-containing 'Zestar' apple (Z; 0.4 mg GAE/mouse/day). Obesity, glucose intolerance, hypertriglyceridemia, and hepatic lipid vacuolation were induced by HF feeding while circulating cholesterol levels remained unchanged. DB apple supplementation did not protect against HF-induced body weight gain, hyperglycemia, hepatic triglyceride level elevation, and hepatic lipid vacuolation at the tested dosage. Future studies should be conducted with increased DB dosage and employ apple (poly)phenols supplemented in the form of extracts or sugar-free powder.
Collapse
Affiliation(s)
- Cindy H J Yu
- Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS, B2N 5E3, Canada
| | - Petra C Kienesberger
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - H P Vasantha Rupasinghe
- Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS, B2N 5E3, Canada.
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
6
|
Lulić AM, Katalinić M. The PNPLA family of enzymes: characterisation and biological role. Arh Hig Rada Toksikol 2023; 74:75-89. [PMID: 37357879 PMCID: PMC10291501 DOI: 10.2478/aiht-2023-74-3723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/01/2023] [Accepted: 05/01/2023] [Indexed: 06/27/2023] Open
Abstract
This paper brings a brief review of the human patatin-like phospholipase domain-containing protein (PNPLA) family. Even though it consists of only nine members, their physiological roles and mechanisms of their catalytic activity are not fully understood. However, the results of a number of knock-out and gain- or loss-of-function research models suggest that these enzymes have an important role in maintaining the homeostasis and integrity of organelle membranes, in cell growth, signalling, cell death, and the metabolism of lipids such as triacylglycerol, phospholipids, ceramides, and retinyl esters. Research has also revealed a connection between PNPLA family member mutations or irregular catalytic activity and the development of various diseases. Here we summarise important findings published so far and discuss their structure, localisation in the cell, distribution in the tissues, specificity for substrates, and their potential physiological role, especially in view of their potential as drug targets.
Collapse
Affiliation(s)
- Ana-Marija Lulić
- Institute for Medical Research and Occupational Health, Biochemistry and Organic Analytical Chemistry Unit, Zagreb, Croatia
| | - Maja Katalinić
- Institute for Medical Research and Occupational Health, Biochemistry and Organic Analytical Chemistry Unit, Zagreb, Croatia
| |
Collapse
|
7
|
Guerrero-Santoro J, Morizane M, Oh SY, Mishima T, Goff JP, Bildirici I, Sadovsky E, Ouyang Y, Tyurin VA, Tyurina YY, Kagan VE, Sadovsky Y. The lipase cofactor CGI58 controls placental lipolysis. JCI Insight 2023; 8:168717. [PMID: 37212279 DOI: 10.1172/jci.insight.168717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/12/2023] [Indexed: 05/23/2023] Open
Abstract
In eutherians, the placenta plays a critical role in the uptake, storage, and metabolism of lipids. These processes govern the availability of fatty acids to the developing fetus, where inadequate supply has been associated with substandard fetal growth. Whereas lipid droplets are essential for the storage of neutral lipids in the placenta and many other tissues, the processes that regulate placental lipid droplet lipolysis remain largely unknown. To assess the role of triglyceride lipases and their cofactors in determining placental lipid droplet and lipid accumulation, we assessed the role of patatin like phospholipase domain containing 2 (PNPLA2) and comparative gene identification-58 (CGI58) in lipid droplet dynamics in the human and mouse placenta. While both proteins are expressed in the placenta, the absence of CGI58, not PNPLA2, markedly increased placental lipid and lipid droplet accumulation. These changes were reversed upon restoration of CGI58 levels selectively in the CGI58-deficient mouse placenta. Using co-immunoprecipitation, we found that, in addition to PNPLA2, PNPLA9 interacts with CGI58. PNPLA9 was dispensable for lipolysis in the mouse placenta yet contributed to lipolysis in human placental trophoblasts. Our findings establish a crucial role for CGI58 in placental lipid droplet dynamics and, by extension, in nutrient supply to the developing fetus.
Collapse
Affiliation(s)
- Jennifer Guerrero-Santoro
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mayumi Morizane
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Soo-Young Oh
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Takuya Mishima
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Julie P Goff
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ibrahim Bildirici
- Department of Obstetrics and Gynecology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Elena Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yingshi Ouyang
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Vladimir A Tyurin
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
| | - Yulia Y Tyurina
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
- Department of Chemistry
- Department of Pharmacology and Chemical Biology
- Department of Radiation Oncology; and
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
8
|
Adipose Triglyceride Lipase in Hepatic Physiology and Pathophysiology. Biomolecules 2021; 12:biom12010057. [PMID: 35053204 PMCID: PMC8773762 DOI: 10.3390/biom12010057] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/28/2021] [Accepted: 12/28/2021] [Indexed: 12/25/2022] Open
Abstract
The liver is extremely active in oxidizing triglycerides (TG) for energy production. An imbalance between TG synthesis and hydrolysis leads to metabolic disorders in the liver, including excessive lipid accumulation, oxidative stress, and ultimately liver damage. Adipose triglyceride lipase (ATGL) is the rate-limiting enzyme that catalyzes the first step of TG breakdown to glycerol and fatty acids. Although its role in controlling lipid homeostasis has been relatively well-studied in the adipose tissue, heart, and skeletal muscle, it remains largely unknown how and to what extent ATGL is regulated in the liver, responds to stimuli and regulators, and mediates disease progression. Therefore, in this review, we describe the current understanding of the structure–function relationship of ATGL, the molecular mechanisms of ATGL regulation at translational and post-translational levels, and—most importantly—its role in lipid and glucose homeostasis in health and disease with a focus on the liver. Advances in understanding the molecular mechanisms underlying hepatic lipid accumulation are crucial to the development of targeted therapies for treating hepatic metabolic disorders.
Collapse
|
9
|
Kanshana JS, Mattila PE, Ewing MC, Wood AN, Schoiswohl G, Meyer AC, Kowalski A, Rosenthal SL, Gingras S, Kaufman BA, Lu R, Weeks DE, McGarvey ST, Minster RL, Hawley NL, Kershaw EE. A murine model of the human CREBRFR457Q obesity-risk variant does not influence energy or glucose homeostasis in response to nutritional stress. PLoS One 2021; 16:e0251895. [PMID: 34520472 PMCID: PMC8439463 DOI: 10.1371/journal.pone.0251895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/09/2021] [Indexed: 01/02/2023] Open
Abstract
Obesity and diabetes have strong heritable components, yet the genetic contributions to these diseases remain largely unexplained. In humans, a missense variant in Creb3 regulatory factor (CREBRF) [rs373863828 (p.Arg457Gln); CREBRFR457Q] is strongly associated with increased odds of obesity but decreased odds of diabetes. Although virtually nothing is known about CREBRF's mechanism of action, emerging evidence implicates it in the adaptive transcriptional response to nutritional stress downstream of TORC1. The objectives of this study were to generate a murine model with knockin of the orthologous variant in mice (CREBRFR458Q) and to test the hypothesis that this CREBRF variant promotes obesity and protects against diabetes by regulating energy and glucose homeostasis downstream of TORC1. To test this hypothesis, we performed extensive phenotypic analysis of CREBRFR458Q knockin mice at baseline and in response to acute (fasting/refeeding), chronic (low- and high-fat diet feeding), and extreme (prolonged fasting) nutritional stress as well as with pharmacological TORC1 inhibition, and aging to 52 weeks. The results demonstrate that the murine CREBRFR458Q model of the human CREBRFR457Q variant does not influence energy/glucose homeostasis in response to these interventions, with the exception of possible greater loss of fat relative to lean mass with age. Alternative preclinical models and/or studies in humans will be required to decipher the mechanisms linking this variant to human health and disease.
Collapse
Affiliation(s)
- Jitendra S. Kanshana
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Polly E. Mattila
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Michael C. Ewing
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Ashlee N. Wood
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Gabriele Schoiswohl
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - Anna C. Meyer
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Aneta Kowalski
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Samantha L. Rosenthal
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Sebastien Gingras
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Brett A. Kaufman
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Ray Lu
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Daniel E. Weeks
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stephen T. McGarvey
- International Health Institute, Department of Epidemiology, Brown University School of Public Health, Providence, Rhode Island, United States of America
- Department of Anthropology, Brown University, Providence, Rhode Island, United States of America
| | - Ryan L. Minster
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Nicola L. Hawley
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Erin E. Kershaw
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
10
|
Li L, Wang J, Li D, Zhang H. Morphine increases myocardial triacylglycerol through regulating adipose triglyceride lipase S406 phosphorylation. Life Sci 2021; 283:119866. [PMID: 34352257 DOI: 10.1016/j.lfs.2021.119866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
AIMS Morphine, a commonly used drug for anesthesia, affects lipid metabolism in different tissues, but the mechanism is currently unclear. Adipose triglyceride lipase (ATGL) is the rate-limiting enzyme responsible for the first step of triglyceride (TG) hydrolysis. Here we aim to investigate whether ATGL phosphorylation is involved in morphine-induced TG accumulation. MAIN METHODS Oil red O staining and TG content analysis were used to detect the effect of morphine on lipid storage. A series of ATGL phosphoamino acid site mutant plasmids were constructed by gene synthesis and transfected to HL-1 cells to evaluate the phosphorylation levels of ATGL phosphoamino acid in morphine-treated HL-1 cells with immunoprecipitation and immunoblotting assay. KEY FINDINGS Morphine acute treatment induced excessive accumulation of TG and decreased the phosphorylation level of ATGL Ser406 in HL-1 cells. Of note, the phosphorylation positive mutation of ATGL Ser406 to aspartic acid effectively reversed morphine-induced excessive accumulation of TG in HL-1 cells. SIGNIFICANCE This discovery will help to fully understand the lipid regulation function of morphine in a new scope. In addition, it will expand the phosphorylation research of ATGL more comprehensively and provide powerful clues for lipid metabolism regulation.
Collapse
Affiliation(s)
- Linghai Li
- Department of Anesthesiology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, 101149, Beijing, China.
| | - Jinghui Wang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, 101149 Beijing, China
| | - Ding Li
- Department of Anesthesiology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, 101149, Beijing, China
| | - Huina Zhang
- Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, 100029 Beijing, China.
| |
Collapse
|
11
|
Chiu YJ, Tu HH, Kung ML, Wu HJ, Chen YW. Fluoxetine ameliorates high-fat diet-induced metabolic abnormalities partially via reduced adipose triglyceride lipase-mediated adipocyte lipolysis. Biomed Pharmacother 2021; 141:111848. [PMID: 34198047 DOI: 10.1016/j.biopha.2021.111848] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/15/2021] [Accepted: 06/24/2021] [Indexed: 02/06/2023] Open
Abstract
Patients with type 2 diabetes mellitus have more risk to develop depression. Fluoxetine (FLX), a selective serotonin reuptake inhibitor (SSRI), is drug for mood and anxiety disorders. Previous studies showed that FLX could induce weight loss in non-depressed clinically overweight individuals. Although the anti-appetite effect of FLX is well-documented, its potential effects on metabolic abnormalities have not been investigated. In this study, we want to investigate whether FLX could be a therapeutic drug against high fat diet (HFD)-induced metabolic disorder. We generated metabolic disorders and depressed mouse model by feeding HFD for 12 weeks at the age of 8 weeks. Then, mice were intraperitoneally injected once daily with FLX (10 mg/kg or 20 mg/kg) for four weeks. Our results showed that FLX alleviated the HFD-induced metabolic dysfunctions and depressive phenotypes in mice. FLX improved systemic glucose homeostasis, at least in part, by improving visceral white adipose tissue (vWAT) insulin signaling. Moreover, FLX reduced circulating plasma leptin level, and decreased the expression of adipose triglyceride lipase (ATGL) and peroxisome proliferator-activated receptor gamma (PPARγ) in vWAT. Our data revealed that FLX also reduced the triglyceride (TG) accumulation in vWAT. Therefore, these findings suggest that FLX exhibits significant potential on comorbidity of metabolic disorder and depression in mice.
Collapse
Affiliation(s)
- Yen-Ju Chiu
- Departments of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ho-Hsiang Tu
- Emergency Department, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi, Taiwan
| | - Mei-Lang Kung
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Hung-Ju Wu
- Departments of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yun-Wen Chen
- Departments of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
12
|
D'Souza K, Acquah C, Mercer A, Paudel Y, Pulinilkunnil T, Udenigwe CC, Kienesberger PC. Whey peptides exacerbate body weight gain and perturb systemic glucose and tissue lipid metabolism in male high-fat fed mice. Food Funct 2021; 12:3552-3561. [PMID: 33900305 DOI: 10.1039/d0fo02610g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Consumption of milk-derived whey proteins has been demonstrated to have insulin-sensitizing effects in mice and humans, in part through the generation of bioactive whey peptides. While whey peptides can prevent insulin resistance in vitro, it is unclear whether consumption of whey peptides can prevent obesity-induced metabolic dysfunction in vivo. We sought to determine whether whey peptides consumption can protect from high fat (HF) diet-induced obesity and dysregulation of glucose homeostasis. Male C57BL/6J mice were fed either a low or HF diet for 13 weeks. HF diet fed mice were provided drinking water with no addition (control), undigested whey protein isolate (WPI, 1 mg ml-1) or whey protein hydrolysate (WPH, 1 mg ml-1) throughout the diet regimen. Mice consuming WPH gained more body weight and were more glucose intolerant compared to those consuming WPI or water only. Despite increased body weight gain, perigonadal adipose tissue weight and lipid accumulation were unchanged. However, excess lipids accumulated ectopically in the liver and skeletal muscle in mice consuming WPH, which was associated with elevated inflammatory markers systemically and in adipose tissue, liver, and skeletal muscle. In skeletal muscle, mitochondrial fat oxidation and electron transport chain proteins were decreased with WPH consumption, indicative of mitochondrial dysfunction. Taken together, our results demonstrate that WPH, but not WPI, exacerbates HF-induced body weight gain and impairs glucose homeostasis, which is accompanied by increased inflammation, ectopic fat accumulation and mitochondrial dysfunction. Thus, our results argue against the use of dietary whey peptide supplementation as a preventative option against HF diet-induced metabolic dysfunction.
Collapse
Affiliation(s)
- Kenneth D'Souza
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick E2L 4L5, Canada.
| | | | | | | | | | | | | |
Collapse
|
13
|
Han SL, Liu Y, Limbu SM, Chen LQ, Zhang ML, Du ZY. The reduction of lipid-sourced energy production caused by ATGL inhibition cannot be compensated by activation of HSL, autophagy, and utilization of other nutrients in fish. FISH PHYSIOLOGY AND BIOCHEMISTRY 2021; 47:173-188. [PMID: 33245450 DOI: 10.1007/s10695-020-00904-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/18/2020] [Indexed: 06/11/2023]
Abstract
The adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL)-mediated lipolysis play important roles in lipid catabolism. ATGL is considered the central rate-limiting enzyme in the mobilization of fatty acids in mammals. Currently, severe fat accumulation has been commonly detected in farmed fish globally. However, the ATGL-mediated lipolysis and the potential synergy among ATGL, HSL, and autophagy, which is another way for lipid breakdown, have not been intensively understood in fish. In the present study, we added Atglistatin as an ATGL-specific inhibitor into the zebrafish diet and fed to the fish for 5 weeks. The results showed that the Atglistatin-treated fish exhibited severe fat deposition, reduced oxygen consumption, and fatty acid β-oxidation, accompanied with increased oxidative stress and inflammation. Furthermore, the Atglistatin-treated fish elevated total and phosphorylation protein expressions of HSL. However, the free fatty acids and lipase activities in organs were still systemically reduced in the Atglistatin-treated fish, and the autophagy marker LC3 was also decreased in the liver. On the other hand, glycogenolysis was stimulated but blood glucose was higher in the Atglistatin-treated fish. The transcriptomic analysis also provided the hint that the protein turnover efficiency in Atglistatin-treated fish was likely to be accelerated, but the protein content in whole fish was not affected. Taken together, ATGL plays crucial roles in energy homeostasis such that its inhibition causes loss of lipid-sourced energy production, which cannot be compensated by activation of HSL, autophagy, and utilization of other nutrients.
Collapse
Affiliation(s)
- Si-Lan Han
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yan Liu
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Samwel M Limbu
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
- Department of Aquatic Sciences and Fisheries Technology, University of Dar es Salaam, P.O. Box 35064, Dar es Salaam, Tanzania
- ECNU-UDSM Joint Research Center for Aquaculture and Fish Biology (JRCAFB), Dar es Salaam, Tanzania
| | - Li-Qiao Chen
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Mei-Ling Zhang
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhen-Yu Du
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China.
- ECNU-UDSM Joint Research Center for Aquaculture and Fish Biology (JRCAFB), Shanghai, China.
| |
Collapse
|
14
|
Prunonosa Cervera I, Gabriel BM, Aldiss P, Morton NM. The phospholipase A2 family's role in metabolic diseases: Focus on skeletal muscle. Physiol Rep 2021; 9:e14662. [PMID: 33433056 PMCID: PMC7802192 DOI: 10.14814/phy2.14662] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022] Open
Abstract
The prevalence of obesity and type 2 diabetes has increased substantially in recent years creating a global health burden. In obesity, skeletal muscle, the main tissue responsible for insulin-mediated glucose uptake, exhibits dysregulation of insulin signaling, glucose uptake, lipid metabolism, and mitochondrial function, thus, promoting type 2 diabetes. The phospholipase A2 (PLA2) enzyme family mediates lipid signaling and membrane remodeling and may play an important role in metabolic disorders such as obesity, diabetes, hyperlipidemia, and fatty liver disease. The PLA2 family consists of 16 members clustered in four groups. PLA2s hydrolyze the sn-2 ester bond of phospholipids generating free fatty acids and lysophospholipids. Differential tissue and subcellular PLA2 expression patterns and the abundance of distinct fatty acyl groups in the target phospholipid determine the impact of individual family members on metabolic functions and, potentially, diseases. Here, we update the current knowledge of the role of the PLA2 family in skeletal muscle, with a view to their potential for therapeutic targeting in metabolic diseases.
Collapse
Affiliation(s)
- Iris Prunonosa Cervera
- Molecular Metabolism GroupCentre for Cardiovascular SciencesQueens Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Brendan M. Gabriel
- Molecular Metabolism GroupCentre for Cardiovascular SciencesQueens Medical Research InstituteUniversity of EdinburghEdinburghUK
- Department of Physiology and PharmacologyIntegrative PhysiologyKarolinska InstituteStockholmSweden
- Aberdeen Cardiovascular & Diabetes CentreThe Rowett InstituteUniversity of AberdeenAberdeenUK
| | - Peter Aldiss
- Molecular Metabolism GroupCentre for Cardiovascular SciencesQueens Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Nicholas M. Morton
- Molecular Metabolism GroupCentre for Cardiovascular SciencesQueens Medical Research InstituteUniversity of EdinburghEdinburghUK
| |
Collapse
|
15
|
Povero D, Johnson SM, Liu J. Hypoxia, hypoxia-inducible gene 2 (HIG2)/HILPDA, and intracellular lipolysis in cancer. Cancer Lett 2020; 493:71-79. [PMID: 32818550 PMCID: PMC11218043 DOI: 10.1016/j.canlet.2020.06.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/27/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022]
Abstract
Tumor tissues are chronically exposed to hypoxia owing to aberrant vascularity. Hypoxia induces metabolic alterations in cancer, thereby promoting aggressive malignancy and metastasis. While previous efforts largely focused on adaptive responses in glucose and glutamine metabolism, recent studies have begun to yield important insight into the hypoxic regulation of lipid metabolic reprogramming in cancer. Emerging evidence points to lipid droplet (LD) accumulation as a hallmark of hypoxic cancer cells. One critical underlying mechanism involves the inhibition of adipose triglyceride lipase (ATGL)-mediated intracellular lipolysis by a small protein encoded by hypoxia-inducible gene 2 (HIG2), also known as hypoxia inducible lipid droplet associated (HILPDA). In this review we summarize and discuss recent key findings on hypoxia-dependent regulation of metabolic adaptations especially lipolysis in cancer. We also pose several questions and hypotheses pertaining to the metabolic impact of lipolytic regulation in cancer under hypoxia and during hypoxia-reoxygenation transition.
Collapse
Affiliation(s)
- Davide Povero
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Division of Endocrinology, Rochester, MN, 55905, USA
| | - Scott M Johnson
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Mayo Clinic College of Medicine & Science, Rochester, MN, 55905, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, 55905, USA
| | - Jun Liu
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Division of Endocrinology, Rochester, MN, 55905, USA.
| |
Collapse
|
16
|
Raje V, Ahern KW, Martinez BA, Howell NL, Oenarto V, Granade ME, Kim JW, Tundup S, Bottermann K, Gödecke A, Keller SR, Kadl A, Bland ML, Harris TE. Adipocyte lipolysis drives acute stress-induced insulin resistance. Sci Rep 2020; 10:18166. [PMID: 33097799 PMCID: PMC7584576 DOI: 10.1038/s41598-020-75321-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Stress hyperglycemia and insulin resistance are evolutionarily conserved metabolic adaptations to severe injury including major trauma, burns, or hemorrhagic shock (HS). In response to injury, the neuroendocrine system increases secretion of counterregulatory hormones that promote rapid mobilization of nutrient stores, impair insulin action, and ultimately cause hyperglycemia, a condition known to impair recovery from injury in the clinical setting. We investigated the contributions of adipocyte lipolysis to the metabolic response to acute stress. Both surgical injury with HS and counterregulatory hormone (epinephrine) infusion profoundly stimulated adipocyte lipolysis and simultaneously triggered insulin resistance and hyperglycemia. When lipolysis was inhibited, the stress-induced insulin resistance and hyperglycemia were largely abolished demonstrating an essential requirement for adipocyte lipolysis in promoting stress-induced insulin resistance. Interestingly, circulating non-esterified fatty acid levels did not increase with lipolysis or correlate with insulin resistance during acute stress. Instead, we show that impaired insulin sensitivity correlated with circulating levels of the adipokine resistin in a lipolysis-dependent manner. Our findings demonstrate the central importance of adipocyte lipolysis in the metabolic response to injury. This insight suggests new approaches to prevent insulin resistance and stress hyperglycemia in trauma and surgery patients and thereby improve outcomes.
Collapse
Affiliation(s)
- Vidisha Raje
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Katelyn W Ahern
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Brittany A Martinez
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Nancy L Howell
- Department of Medicine, Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| | - Vici Oenarto
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA.,Institute of Cardiovascular Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Mitchell E Granade
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Jae Woo Kim
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Smanla Tundup
- Department of Medicine, Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VA, USA
| | | | - Axel Gödecke
- Institute of Cardiovascular Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Susanna R Keller
- Department of Medicine, Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| | - Alexandra Kadl
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA.,Department of Medicine, Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VA, USA
| | - Michelle L Bland
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Thurl E Harris
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
17
|
Biswas D, Dao KT, Mercer A, Cowie AM, Duffley L, El Hiani Y, Kienesberger PC, Pulinilkunnil T. Branched-chain ketoacid overload inhibits insulin action in the muscle. J Biol Chem 2020; 295:15597-15621. [PMID: 32878988 DOI: 10.1074/jbc.ra120.013121] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 08/29/2020] [Indexed: 12/18/2022] Open
Abstract
Branched-chain α-keto acids (BCKAs) are catabolites of branched-chain amino acids (BCAAs). Intracellular BCKAs are cleared by branched-chain ketoacid dehydrogenase (BCKDH), which is sensitive to inhibitory phosphorylation by BCKD kinase (BCKDK). Accumulation of BCKAs is an indicator of defective BCAA catabolism and has been correlated with glucose intolerance and cardiac dysfunction. However, it is unclear whether BCKAs directly alter insulin signaling and function in the skeletal and cardiac muscle cell. Furthermore, the role of excess fatty acids (FAs) in perturbing BCAA catabolism and BCKA availability merits investigation. By using immunoblotting and ultra-performance liquid chromatography MS/MS to analyze the hearts of fasted mice, we observed decreased BCAA-catabolizing enzyme expression and increased circulating BCKAs, but not BCAAs. In mice subjected to diet-induced obesity (DIO), we observed similar increases in circulating BCKAs with concomitant changes in BCAA-catabolizing enzyme expression only in the skeletal muscle. Effects of DIO were recapitulated by simulating lipotoxicity in skeletal muscle cells treated with saturated FA, palmitate. Exposure of muscle cells to high concentrations of BCKAs resulted in inhibition of insulin-induced AKT phosphorylation, decreased glucose uptake, and mitochondrial oxygen consumption. Altering intracellular clearance of BCKAs by genetic modulation of BCKDK and BCKDHA expression showed similar effects on AKT phosphorylation. BCKAs increased protein translation and mTORC1 activation. Pretreating cells with mTORC1 inhibitor rapamycin restored BCKA's effect on insulin-induced AKT phosphorylation. This study provides evidence for FA-mediated regulation of BCAA-catabolizing enzymes and BCKA content and highlights the biological role of BCKAs in regulating muscle insulin signaling and function.
Collapse
Affiliation(s)
- Dipsikha Biswas
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Khoi T Dao
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Angella Mercer
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Andrew M Cowie
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Luke Duffley
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Yassine El Hiani
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Petra C Kienesberger
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada.
| |
Collapse
|
18
|
Llanos AAM, Lin Y, Chen W, Yao S, Norin J, Chekmareva MA, Omene C, Cong L, Omilian AR, Khoury T, Hong CC, Ganesan S, Foran DJ, Higgins M, Ambrosone CB, Bandera EV, Demissie K. Immunohistochemical analysis of adipokine and adipokine receptor expression in the breast tumor microenvironment: associations of lower leptin receptor expression with estrogen receptor-negative status and triple-negative subtype. Breast Cancer Res 2020; 22:18. [PMID: 32046756 PMCID: PMC7014630 DOI: 10.1186/s13058-020-1256-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/29/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The molecular mechanisms underlying the association between increased adiposity and aggressive breast cancer phenotypes remain unclear, but likely involve the adipokines, leptin (LEP) and adiponectin (ADIPOQ), and their receptors (LEPR, ADIPOR1, ADIPOR2). METHODS We used immunohistochemistry (IHC) to assess LEP, LEPR, ADIPOQ, ADIPOR1, and ADIPOR2 expression in breast tumor tissue microarrays among a sample of 720 women recently diagnosed with breast cancer (540 of whom self-identified as Black). We scored IHC expression quantitatively, using digital pathology analysis. We abstracted data on tumor grade, tumor size, tumor stage, lymph node status, Ki67, estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) from pathology records, and used ER, PR, and HER2 expression data to classify breast cancer subtype. We used multivariable mixed effects models to estimate associations of IHC expression with tumor clinicopathology, in the overall sample and separately among Blacks. RESULTS Larger proportions of Black than White women were overweight or obese and had more aggressive tumor features. Older age, Black race, postmenopausal status, and higher body mass index were associated with higher LEPR IHC expression. In multivariable models, lower LEPR IHC expression was associated with ER-negative status and triple-negative subtype (P < 0.0001) in the overall sample and among Black women only. LEP, ADIPOQ, ADIPOR1, and ADIPOR2 IHC expression were not significantly associated with breast tumor clinicopathology. CONCLUSIONS Lower LEPR IHC expression within the breast tumor microenvironment might contribute mechanistically to inter-individual variation in aggressive breast cancer clinicopathology, particularly ER-negative status and triple-negative subtype.
Collapse
Affiliation(s)
- Adana A M Llanos
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, USA. .,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.
| | - Yong Lin
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Wenjin Chen
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Song Yao
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jorden Norin
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Cell Biology and Neuroscience, Rutgers School of Arts and Sciences, New Brunswick, NJ, USA
| | - Marina A Chekmareva
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Coral Omene
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Medicine, Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Lei Cong
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Angela R Omilian
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Thaer Khoury
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Chi-Chen Hong
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Shridar Ganesan
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Medicine, Robert Wood Johnson Medical School, New Brunswick, NJ, USA.,Department of Pharmacology, Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - David J Foran
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Michael Higgins
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Christine B Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Elisa V Bandera
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Kitaw Demissie
- Department of Epidemiology and Biostatistics, SUNY Downstate Health Sciences University School of Public Health, Brooklyn, NY, USA
| |
Collapse
|
19
|
Yu L, Li Y, Grisé A, Wang H. CGI-58: Versatile Regulator of Intracellular Lipid Droplet Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:197-222. [PMID: 32705602 PMCID: PMC8063591 DOI: 10.1007/978-981-15-6082-8_13] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Comparative gene identification-58 (CGI-58), also known as α/β-hydrolase domain-containing 5 (ABHD5), is a member of a large family of proteins containing an α/β-hydrolase-fold. CGI-58 is well-known as the co-activator of adipose triglyceride lipase (ATGL), which is a key enzyme initiating cytosolic lipid droplet lipolysis. Mutations in either the human CGI-58 or ATGL gene cause an autosomal recessive neutral lipid storage disease, characterized by the excessive accumulation of triglyceride (TAG)-rich lipid droplets in the cytoplasm of almost all cell types. CGI-58, however, has ATGL-independent functions. Distinct phenotypes associated with CGI-58 deficiency commonly include ichthyosis (scaly dry skin), nonalcoholic steatohepatitis, and hepatic fibrosis. Through regulated interactions with multiple protein families, CGI-58 controls many metabolic and signaling pathways, such as lipid and glucose metabolism, energy balance, insulin signaling, inflammatory responses, and thermogenesis. Recent studies have shown that CGI-58 regulates the pathogenesis of common metabolic diseases in a tissue-specific manner. Future studies are needed to molecularly define ATGL-independent functions of CGI-58, including the newly identified serine protease activity of CGI-58. Elucidation of these versatile functions of CGI-58 may uncover fundamental cellular processes governing lipid and energy homeostasis, which may help develop novel approaches that counter against obesity and its associated metabolic sequelae.
Collapse
Affiliation(s)
- Liqing Yu
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Yi Li
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alison Grisé
- College of Computer, Math, and Natural Sciences, College of Behavioral and Social Sciences, University of Maryland, College Park, MD, USA
| | - Huan Wang
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
20
|
Trites MJ, Clugston RD. The role of adipose triglyceride lipase in lipid and glucose homeostasis: lessons from transgenic mice. Lipids Health Dis 2019; 18:204. [PMID: 31757217 PMCID: PMC6874817 DOI: 10.1186/s12944-019-1151-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/07/2019] [Indexed: 12/14/2022] Open
Abstract
The ability of mammals to store and draw on fat reserves has been a driving force throughout evolution in an environment with intermittent nutrient availability. The discovery of adipose triglyceride lipase (ATGL) as a triglyceride lipase provided a heightened understanding of the mechanisms governing mobilization of fat reserves from adipose tissue. ATGL catalyses the initial step in adipose triglyceride lipolysis, working in concert with other enzymes to mobilize triglyceride for energy production. In addition to the role of ATGL in adipose tissue triglyceride mobilization, ATGL plays crucial roles in regulating lipid homeostasis in other tissues. These roles have been characterized primarily using transgenic mice with tissue-specific ATGL ablation. For example, the global ATGL knockout induces a severe cardiac defect that results in premature mortality that is mimicked by inducible cardiomyocyte-specific ATGL knockout. Global- and adipose-specific ATGL ablation induces a whole-body shift from lipid metabolism to glucose metabolism to satisfy metabolic demand primarily facilitated by an increase in glucose uptake by skeletal muscle. Generation of liver-specific ATGL knockouts has implicated hepatic lipolysis as a critical component of normal liver function. Analysis of β-cell ATGL knockouts implicates the necessity of pancreatic ATGL in insulin secretion. The objective of this review is to discuss the contributions of ATGL to systemic lipid- and glucose-homeostasis discovered through the study of transgenic mice.
Collapse
Affiliation(s)
- Michael J Trites
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada.,Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Robin D Clugston
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada. .,Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
21
|
Cipolletta E, Gambardella J, Fiordelisi A, Del Giudice C, Di Vaia E, Ciccarelli M, Sala M, Campiglia P, Coscioni E, Trimarco B, Sorriento D, Iaccarino G. Antidiabetic and Cardioprotective Effects of Pharmacological Inhibition of GRK2 in db/db Mice. Int J Mol Sci 2019; 20:ijms20061492. [PMID: 30934608 PMCID: PMC6470575 DOI: 10.3390/ijms20061492] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/17/2019] [Accepted: 03/20/2019] [Indexed: 12/17/2022] Open
Abstract
Despite the availability of several therapies for the management of blood glucose in diabetic patients, most of the treatments do not show benefits on diabetic cardiomyopathy, while others even favor the progression of the disease. New pharmacological targets are needed that might help the management of diabetes and its cardiovascular complications at the same time. GRK2 appears a promising target, given its established role in insulin resistance and in systolic heart failure. Using a custom peptide inhibitor of GRK2, we assessed in vitro in L6 myoblasts the effects of GRK2 inhibition on glucose extraction and insulin signaling. Afterwards, we treated diabetic male mice (db/db) for 2 weeks. Glucose tolerance (IGTT) and insulin sensitivity (ITT) were ameliorated, as was skeletal muscle glucose uptake and insulin signaling. In the heart, at the same time, the GRK2 inhibitor ameliorated inflammatory and cytokine responses, reduced oxidative stress, and corrected patterns of fetal gene expression, typical of diabetic cardiomyopathy. GRK2 inhibition represents a promising therapeutic target for diabetes and its cardiovascular complications.
Collapse
Affiliation(s)
- Ersilia Cipolletta
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, 80131 Napoli, Italy.
| | - Jessica Gambardella
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, 80131 Napoli, Italy.
| | - Antonella Fiordelisi
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, 80131 Napoli, Italy.
| | - Carmine Del Giudice
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, 80131 Napoli, Italy.
| | - Eugenio Di Vaia
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, 80131 Napoli, Italy.
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy.
| | - Marina Sala
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy.
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy.
| | - Enrico Coscioni
- AOU San Giovanni di Dio e Ruggi d'Aragona, 84131 Salerno, Italy.
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, 80131 Napoli, Italy.
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, 80131 Napoli, Italy.
| | - Guido Iaccarino
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, 80131 Napoli, Italy.
| |
Collapse
|
22
|
Defective fasting-induced PKA activation impairs adipose tissue glycogen degradation in obese Zucker rats. Int J Obes (Lond) 2019; 44:500-509. [PMID: 30705392 DOI: 10.1038/s41366-019-0327-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/20/2018] [Accepted: 01/11/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Obesity is associated with development of insulin resistance in adipose tissue (AT). Human obesity has been associated with increased glycogen deposition in adipocytes. Adipocytes synthesise glycogen prior to the formation of lipids. The present study examined adipose glycogen content in obese Zucker rats and the effect of fasting on glycogen-metabolising enzymes. We hypothesised that obesity imposes a blunted response to fasting through impaired activation of glycogen-metabolizing enzymes, which dampens glycogen mobilization in obese Zucker rats. METHODS We investigated the effect of 24h fasting on AT glycogen metabolism in 12-week old obese Zucker rats. Epididymal fat pads were collected from rats fed ad-libitum and fasted for 24h. Glycogen content, glycogen synthase and phosphorylase enzyme activity, and PKA activity were analysed as well as total and phosphorylated protein content for glycogen-metabolizing enzymes glycogen synthase and phosphorylase, glucose transporter GLUT4, and cAMP-dependent response element binding protein levels. RESULTS Twelve-week old obese Zucker rats showed increased AT glycogen content (adipose glycogen content [mean ± SD], lean: 3.95 ± 2.78 to 0.75 + 0.69 µg.mg-1; p < 0.005 fed vs fasted, and obese: 5.23 ± 3.38 to 5.019 ± 1.99 µg.mg-1; p = ns fed and fasted and p < 0.005 lean vs obese), and impaired fasting-induced glycogen mobilization following a 24h fast. These defects were associated with dysfunctional glycogen-metabolizing enzymes, characterized by: (1) blunted phosphorylation-mediated activation and downregulated protein expression of glycogen phosphorylase, and (2) an impaired phosphorylation-mediated inactivation of glycogen synthase. Furthermore, these defects were related to impaired fasting-induced protein kinase A (PKA) activation. CONCLUSION This study provides evidence of a defective glycogen metabolism in the adipose associated with impaired fasting-induced activation of the upstream kinase protein kinase A, which render a converging point to obesity-related primary alterations in carbohydrate and lipid metabolism in the AT.
Collapse
|
23
|
Of mice and men: The physiological role of adipose triglyceride lipase (ATGL). Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:880-899. [PMID: 30367950 PMCID: PMC6439276 DOI: 10.1016/j.bbalip.2018.10.008] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/12/2022]
Abstract
Adipose triglyceride lipase (ATGL) has been discovered 14 years ago and revised our view on intracellular triglyceride (TG) mobilization – a process termed lipolysis. ATGL initiates the hydrolysis of TGs to release fatty acids (FAs) that are crucial energy substrates, precursors for the synthesis of membrane lipids, and ligands of nuclear receptors. Thus, ATGL is a key enzyme in whole-body energy homeostasis. In this review, we give an update on how ATGL is regulated on the transcriptional and post-transcriptional level and how this affects the enzymes' activity in the context of neutral lipid catabolism. In depth, we highlight and discuss the numerous physiological functions of ATGL in lipid and energy metabolism. Over more than a decade, different genetic mouse models lacking or overexpressing ATGL in a cell- or tissue-specific manner have been generated and characterized. Moreover, pharmacological studies became available due to the development of a specific murine ATGL inhibitor (Atglistatin®). The identification of patients with mutations in the human gene encoding ATGL and their disease spectrum has underpinned the importance of ATGL in humans. Together, mouse models and human data have advanced our understanding of the physiological role of ATGL in lipid and energy metabolism in adipose and non-adipose tissues, and of the pathophysiological consequences of ATGL dysfunction in mice and men. Summary of mouse models with genetic or pharmacological manipulation of ATGL. Summary of patients with mutations in the human gene encoding ATGL. In depth discussion of the role of ATGL in numerous physiological processes in mice and men.
Collapse
|
24
|
Petan T, Jarc E, Jusović M. Lipid Droplets in Cancer: Guardians of Fat in a Stressful World. Molecules 2018; 23:molecules23081941. [PMID: 30081476 PMCID: PMC6222695 DOI: 10.3390/molecules23081941] [Citation(s) in RCA: 248] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 12/12/2022] Open
Abstract
Cancer cells possess remarkable abilities to adapt to adverse environmental conditions. Their survival during severe nutrient and oxidative stress depends on their capacity to acquire extracellular lipids and the plasticity of their mechanisms for intracellular lipid synthesis, mobilisation, and recycling. Lipid droplets, cytosolic fat storage organelles present in most cells from yeast to men, are emerging as major regulators of lipid metabolism, trafficking, and signalling in various cells and tissues exposed to stress. Their biogenesis is induced by nutrient and oxidative stress and they accumulate in various cancers. Lipid droplets act as switches that coordinate lipid trafficking and consumption for different purposes in the cell, such as energy production, protection against oxidative stress or membrane biogenesis during rapid cell growth. They sequester toxic lipids, such as fatty acids, cholesterol and ceramides, thereby preventing lipotoxic cell damage and engage in a complex relationship with autophagy. Here, we focus on the emerging mechanisms of stress-induced lipid droplet biogenesis; their roles during nutrient, lipotoxic, and oxidative stress; and the relationship between lipid droplets and autophagy. The recently discovered principles of lipid droplet biology can improve our understanding of the mechanisms that govern cancer cell adaptability and resilience to stress.
Collapse
Affiliation(s)
- Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana SI-1000, Slovenia.
| | - Eva Jarc
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana SI-1000, Slovenia.
- Jožef Stefan International Postgraduate School, Ljubljana SI-1000, Slovenia.
| | - Maida Jusović
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana SI-1000, Slovenia.
- Jožef Stefan International Postgraduate School, Ljubljana SI-1000, Slovenia.
| |
Collapse
|
25
|
D'Souza K, Nzirorera C, Cowie AM, Varghese GP, Trivedi P, Eichmann TO, Biswas D, Touaibia M, Morris AJ, Aidinis V, Kane DA, Pulinilkunnil T, Kienesberger PC. Autotaxin-LPA signaling contributes to obesity-induced insulin resistance in muscle and impairs mitochondrial metabolism. J Lipid Res 2018; 59:1805-1817. [PMID: 30072447 DOI: 10.1194/jlr.m082008] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 06/26/2018] [Indexed: 01/14/2023] Open
Abstract
Autotaxin (ATX) is an adipokine that generates the bioactive lipid, lysophosphatidic acid (LPA). ATX-LPA signaling has been implicated in diet-induced obesity and systemic insulin resistance. However, it remains unclear whether the ATX-LPA pathway influences insulin function and energy metabolism in target tissues, particularly skeletal muscle, the major site of insulin-stimulated glucose disposal. The objective of this study was to test whether the ATX-LPA pathway impacts tissue insulin signaling and mitochondrial metabolism in skeletal muscle during obesity. Male mice with heterozygous ATX deficiency (ATX+/-) were protected from obesity, systemic insulin resistance, and cardiomyocyte dysfunction following high-fat high-sucrose (HFHS) feeding. HFHS-fed ATX+/- mice also had improved insulin-stimulated AKT phosphorylation in white adipose tissue, liver, heart, and skeletal muscle. Preserved insulin-stimulated glucose transport in muscle from HFHS-fed ATX+/- mice was associated with improved mitochondrial pyruvate oxidation in the absence of changes in fat oxidation and ectopic lipid accumulation. Similarly, incubation with LPA decreased insulin-stimulated AKT phosphorylation and mitochondrial energy metabolism in C2C12 myotubes at baseline and following palmitate-induced insulin resistance. Taken together, our results suggest that the ATX-LPA pathway contributes to obesity-induced insulin resistance in metabolically relevant tissues. Our data also suggest that LPA directly impairs skeletal muscle insulin signaling and mitochondrial function.
Collapse
Affiliation(s)
- Kenneth D'Souza
- Dalhousie Medicine New Brunswick, Department of Biochemistry and Molecular Biology, Dalhousie University, Saint John, New Brunswick E2L 4L5, Canada
| | - Carine Nzirorera
- Dalhousie Medicine New Brunswick, Department of Biochemistry and Molecular Biology, Dalhousie University, Saint John, New Brunswick E2L 4L5, Canada
| | - Andrew M Cowie
- Dalhousie Medicine New Brunswick, Department of Biochemistry and Molecular Biology, Dalhousie University, Saint John, New Brunswick E2L 4L5, Canada
| | - Geena P Varghese
- Dalhousie Medicine New Brunswick, Department of Biochemistry and Molecular Biology, Dalhousie University, Saint John, New Brunswick E2L 4L5, Canada
| | - Purvi Trivedi
- Dalhousie Medicine New Brunswick, Department of Biochemistry and Molecular Biology, Dalhousie University, Saint John, New Brunswick E2L 4L5, Canada
| | - Thomas O Eichmann
- Institute of Molecular Biosciences, University of Graz and Center for Explorative Lipidomics, BioTechMed-Graz, 8010 Graz, Austria
| | - Dipsikha Biswas
- Dalhousie Medicine New Brunswick, Department of Biochemistry and Molecular Biology, Dalhousie University, Saint John, New Brunswick E2L 4L5, Canada
| | - Mohamed Touaibia
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick E1A 3E9, Canada
| | - Andrew J Morris
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY 40536 and Lexington Veterans Affairs Medical Center, Lexington, KY 40511
| | - Vassilis Aidinis
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", 16672 Athens, Greece
| | - Daniel A Kane
- Department of Human Kinetics, St. Francis Xavier University, Antigonish, Nova Scotia B2G 2W5, Canada
| | - Thomas Pulinilkunnil
- Dalhousie Medicine New Brunswick, Department of Biochemistry and Molecular Biology, Dalhousie University, Saint John, New Brunswick E2L 4L5, Canada
| | - Petra C Kienesberger
- Dalhousie Medicine New Brunswick, Department of Biochemistry and Molecular Biology, Dalhousie University, Saint John, New Brunswick E2L 4L5, Canada
| |
Collapse
|
26
|
Nakano T, Seino K, Wakabayashi I, Stafforini DM, Topham MK, Goto K. Deletion of diacylglycerol kinase ε confers susceptibility to obesity
via
reduced lipolytic activity in murine adipocytes. FASEB J 2018; 32:4121-4131. [DOI: 10.1096/fj.201701050r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Tomoyuki Nakano
- Department of Anatomy and Cell BiologyYamagata University School of MedicineYamagataJapan
| | - Keiko Seino
- Department of Anatomy and Cell BiologyYamagata University School of MedicineYamagataJapan
| | - Ichiro Wakabayashi
- Department of Environmental and Preventive MedicineHyogo College of MedicineNishinomiyaHyogoJapan
| | | | | | - Kaoru Goto
- Department of Anatomy and Cell BiologyYamagata University School of MedicineYamagataJapan
| |
Collapse
|
27
|
Hints on ATGL implications in cancer: beyond bioenergetic clues. Cell Death Dis 2018; 9:316. [PMID: 29472527 PMCID: PMC5833653 DOI: 10.1038/s41419-018-0345-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 12/21/2022]
Abstract
Among metabolic rearrangements occurring in cancer cells, lipid metabolism alteration has become a hallmark, aimed at sustaining accelerated proliferation. In particular, fatty acids (FAs) are dramatically required by cancer cells as signalling molecules and membrane building blocks, beyond bioenergetics. Along with de novo biosynthesis, free FAs derive from dietary sources or from intracellular lipid droplets, which represent the storage of triacylglycerols (TAGs). Adipose triglyceride lipase (ATGL) is the rate-limiting enzyme of lipolysis, catalysing the first step of intracellular TAGs hydrolysis in several tissues. However, the roles of ATGL in cancer are still neglected though a putative tumour suppressor function of ATGL has been envisaged, as its expression is frequently reduced in different human cancers (e.g., lung, muscle, and pancreas). In this review, we will introduce lipid metabolism focusing on ATGL functions and regulation in normal cell physiology providing also speculative perspectives on potential non-energetic functions of ATGL in cancer. In particular, we will discuss how ATGL is implicated, mainly through the peroxisome proliferator-activated receptor-α (PPAR-α) signalling, in inflammation, redox homoeostasis and autophagy, which are well-known processes deregulated during cancer formation and/or progression.
Collapse
|
28
|
Li X, Sun K. Regulation of Lipolysis in Adipose Tissue and Clinical Significance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1090:199-210. [PMID: 30390292 DOI: 10.1007/978-981-13-1286-1_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Lipolysis is a critical process to hydrolyze triglyceride in adipose tissue, thereby breaking down the stored lipid and maintaining energy homeostasis. Recent studies have made significant progress in understanding the steps of lipolysis. This chapter discusses the major pathways that regulate lipolysis in adipose tissue. Specifically we focus on the mechanisms by which the activities of critical lipolytic enzymes are regulated. We further discuss how the lipolysis is regulated by other factors, including insulin and neurotransmitters, in particular catecholamines and the role of sympathetic nervous system in the whole process. Finally we provide clinical perspectives about the novel therapeutic strategies to target or promote adipose tissue lipolysis for treatment/prevention of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Xin Li
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kai Sun
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
29
|
Insulin action and resistance in obesity and type 2 diabetes. Nat Med 2017; 23:804-814. [PMID: 28697184 DOI: 10.1038/nm.4350] [Citation(s) in RCA: 838] [Impact Index Per Article: 104.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 05/11/2017] [Indexed: 12/12/2022]
Abstract
Nutritional excess is a major forerunner of type 2 diabetes. It enhances the secretion of insulin, but attenuates insulin's metabolic actions in the liver, skeletal muscle and adipose tissue. However, conflicting evidence indicates a lack of knowledge of the timing of these events during the development of obesity and diabetes, pointing to a key gap in our understanding of metabolic disease. This Perspective reviews alternate viewpoints and recent results on the temporal and mechanistic connections between hyperinsulinemia, obesity and insulin resistance. Although much attention has addressed early steps in the insulin signaling cascade, insulin resistance in obesity seems to be largely elicited downstream of these steps. New findings also connect insulin resistance to extensive metabolic cross-talk between the liver, adipose tissue, pancreas and skeletal muscle. These and other advances over the past 5 years offer exciting opportunities and daunting challenges for the development of new therapeutic strategies for the treatment of type 2 diabetes.
Collapse
|
30
|
Critical roles for α/β hydrolase domain 5 (ABHD5)/comparative gene identification-58 (CGI-58) at the lipid droplet interface and beyond. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1233-1241. [PMID: 28827091 DOI: 10.1016/j.bbalip.2017.07.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 07/24/2017] [Accepted: 07/31/2017] [Indexed: 01/04/2023]
Abstract
Mutations in the gene encoding comparative gene identification 58 (CGI-58), also known as α β hydrolase domain-containing 5 (ABHD5), cause neutral lipid storage disorder with ichthyosis (NLSDI). This inborn error in metabolism is characterized by ectopic accumulation of triacylglycerols (TAG) within cytoplasmic lipid droplets in multiple cell types. Studies over the past decade have clearly demonstrated that CGI-58 is a potent regulator of TAG hydrolysis in the disease-relevant cell types. However, despite the reproducible genetic link between CGI-58 mutations and TAG storage, the molecular mechanisms by which CGI-58 regulates TAG hydrolysis are still incompletely understood. It is clear that CGI-58 can regulate TAG hydrolysis by activating the major TAG hydrolase adipose triglyceride lipase (ATGL), yet CGI-58 can also regulate lipid metabolism via mechanisms that do not involve ATGL. This review highlights recent progress made in defining the physiologic and biochemical function of CGI-58, and its broader role in energy homeostasis. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
|
31
|
Jin J, Huang S, Wang L, Leng Y, Lu W. Design and synthesis of Atglistatin derivatives as adipose triglyceride lipase inhibitors. Chem Biol Drug Des 2017; 90:1122-1133. [DOI: 10.1111/cbdd.13029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/18/2017] [Accepted: 05/11/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Jiyu Jin
- School of Chemistry and Molecular Engineering; East China Normal University; Shanghai China
| | - Suling Huang
- State Key Laboratory of Drug Research; Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Lei Wang
- School of Chemistry and Molecular Engineering; East China Normal University; Shanghai China
| | - Ying Leng
- State Key Laboratory of Drug Research; Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Wei Lu
- School of Chemistry and Molecular Engineering; East China Normal University; Shanghai China
| |
Collapse
|
32
|
Zhang X, Heckmann BL, Campbell LE, Liu J. G0S2: A small giant controller of lipolysis and adipose-liver fatty acid flux. Biochim Biophys Acta Mol Cell Biol Lipids 2017. [PMID: 28645852 DOI: 10.1016/j.bbalip.2017.06.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The discovery of adipose triglyceride lipase (ATGL) and its coactivator comparative gene identification-58 (CGI-58) provided a major paradigm shift in the understanding of intracellular lipolysis in both adipocytes and nonadipocyte cells. The subsequent discovery of G0/G1 switch gene 2 (G0S2) as a potent endogenous inhibitor of ATGL revealed a unique mechanism governing lipolysis and fatty acid (FA) availability. G0S2 is highly conserved in vertebrates, and exhibits cyclical expression pattern between adipose tissue and liver that is critical to lipid flux and energy homeostasis in these two tissues. Biochemical and cell biological studies have demonstrated that a direct interaction with ATGL mediates G0S2's inhibitory effects on lipolysis and lipid droplet degradation. In this review we examine evidence obtained from recent in vitro and in vivo studies that lends support to the proof-of-principle concept that G0S2 functions as a master regulator of tissue-specific balance of TG storage vs. mobilization, partitioning of metabolic fuels between adipose and liver, and the whole-body adaptive energy response. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, AZ, United States; HEAL(th) Program, Mayo Clinic, Scottsdale, AZ, United States
| | - Bradlee L Heckmann
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Latoya E Campbell
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Jun Liu
- Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, AZ, United States; HEAL(th) Program, Mayo Clinic, Scottsdale, AZ, United States; Division of Endocrinology, Mayo Clinic, Scottsdale, AZ, United States.
| |
Collapse
|
33
|
Natarajan SK, Rasineni K, Ganesan M, Feng D, McVicker BL, McNiven MA, Osna NA, Mott JL, Casey CA, Kharbanda KK. Structure, Function and Metabolism of Hepatic and Adipose Tissue Lipid Droplets: Implications in Alcoholic Liver Disease. Curr Mol Pharmacol 2017; 10:237-248. [PMID: 26278390 PMCID: PMC4820363 DOI: 10.2174/1874467208666150817111727] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 08/07/2015] [Accepted: 08/07/2015] [Indexed: 02/08/2023]
Abstract
For more than 30 years, lipid droplets (LDs) were considered as an inert bag of lipid for storage of energy-rich fat molecules. Following a paradigm shift almost a decade ago, LDs are presently considered an active subcellular organelle especially designed for assembling, storing and subsequently supplying lipids for generating energy and membrane synthesis (and in the case of hepatocytes for VLDL secretion). LDs also play a central role in many other cellular functions such as viral assembly and protein degradation. Here, we have explored the structural and functional changes that occur in hepatic and adipose tissue LDs following chronic ethanol consumption in relation to their role in the pathogenesis of alcoholic liver injury.
Collapse
Affiliation(s)
- Sathish Kumar Natarajan
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center
| | - Karuna Rasineni
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Murali Ganesan
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Dan Feng
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Benita L. McVicker
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Mark A. McNiven
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Natalia A. Osna
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Justin L. Mott
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center
| | - Carol A. Casey
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center
| | - Kusum K. Kharbanda
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center
| |
Collapse
|
34
|
Lipases and their inhibitors in health and disease. Chem Biol Interact 2016; 259:211-222. [DOI: 10.1016/j.cbi.2016.04.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/04/2016] [Accepted: 04/04/2016] [Indexed: 02/07/2023]
|
35
|
Chen G, Yu D, Nian X, Liu J, Koenig RJ, Xu B, Sheng L. LncRNA SRA promotes hepatic steatosis through repressing the expression of adipose triglyceride lipase (ATGL). Sci Rep 2016; 6:35531. [PMID: 27759039 PMCID: PMC5069493 DOI: 10.1038/srep35531] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 09/30/2016] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), the most common form of chronic liver disease, manifests as an over-accumulation of hepatic fat. We have recently shown that mice with genetic knockout of a long non-coding RNA (lncRNA) steroid receptor RNA activator (SRA) (SRAKO) are resistant to high fat diet-induced obesity with a phenotype that includes improved glucose tolerance and attenuated hepatic steatosis. The underlying mechanism was investigated in the present study. We found that hepatic levels of SRA and adipose triglyceride lipase (ATGL), a major hepatic triacylglycerol (TAG) hydrolase, were inversely regulated by fasting in mice, and the expression of liver ATGL was induced by SRAKO under normal and high fat diet (HFD) feeding. Loss of SRA in primary hepatocytes or a hepatocyte cell line upregulates, but forced expression of SRA inhibits ATGL expression and free fatty acids (FFA) β-oxidation. SRA inhibits ATGL promoter activity, primarily by inhibiting the otherwise-inductive effects of the transcription factor, forkhead box protein O1 (FoxO1). Our data reveal a novel function of SRA in promoting hepatic steatosis through repression of ATGL expression.
Collapse
Affiliation(s)
- Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Dongsheng Yu
- Department of Pharmacology, School of Basic Medical Science, Nanjing Medical University, 140 Hanzhong Rd., Nanjing, Jiangsu, 210029, China
| | - Xue Nian
- Department of Pharmacology, School of Basic Medical Science, Nanjing Medical University, 140 Hanzhong Rd., Nanjing, Jiangsu, 210029, China
| | - Junyi Liu
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ronald J Koenig
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, MI 48109-5678, USA
| | - Bin Xu
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, MI 48109-5678, USA
| | - Liang Sheng
- Department of Pharmacology, School of Basic Medical Science, Nanjing Medical University, 140 Hanzhong Rd., Nanjing, Jiangsu, 210029, China
| |
Collapse
|
36
|
Roy PP, D'Souza K, Cuperlovic-Culf M, Kienesberger PC, Touaibia M. New Atglistatin closely related analogues: Synthesis and structure-activity relationship towards adipose triglyceride lipase inhibition. Eur J Med Chem 2016; 118:290-8. [DOI: 10.1016/j.ejmech.2016.04.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 04/06/2016] [Accepted: 04/07/2016] [Indexed: 11/28/2022]
|
37
|
Lord CC, Ferguson D, Thomas G, Brown AL, Schugar RC, Burrows A, Gromovsky AD, Betters J, Neumann C, Sacks J, Marshall S, Watts R, Schweiger M, Lee RG, Crooke RM, Graham MJ, Lathia JD, Sakaguchi TF, Lehner R, Haemmerle G, Zechner R, Brown JM. Regulation of Hepatic Triacylglycerol Metabolism by CGI-58 Does Not Require ATGL Co-activation. Cell Rep 2016; 16:939-949. [PMID: 27396333 DOI: 10.1016/j.celrep.2016.06.049] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/20/2016] [Accepted: 06/10/2016] [Indexed: 01/23/2023] Open
Abstract
Adipose triglyceride lipase (ATGL) and comparative gene identification 58 (CGI-58) are critical regulators of triacylglycerol (TAG) turnover. CGI-58 is thought to regulate TAG mobilization by stimulating the enzymatic activity of ATGL. However, it is not known whether this coactivation function of CGI-58 occurs in vivo. Moreover, the phenotype of human CGI-58 mutations suggests ATGL-independent functions. Through direct comparison of mice with single or double deficiency of CGI-58 and ATGL, we show here that CGI-58 knockdown causes hepatic steatosis in both the presence and absence of ATGL. CGI-58 also regulates hepatic diacylglycerol (DAG) and inflammation in an ATGL-independent manner. Interestingly, ATGL deficiency, but not CGI-58 deficiency, results in suppression of the hepatic and adipose de novo lipogenic program. Collectively, these findings show that CGI-58 regulates hepatic neutral lipid storage and inflammation in the genetic absence of ATGL, demonstrating that mechanisms driving TAG lipolysis in hepatocytes differ significantly from those in adipocytes.
Collapse
Affiliation(s)
- Caleb C Lord
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA; Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9077, USA
| | - Daniel Ferguson
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA; Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Gwynneth Thomas
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA
| | - Amanda L Brown
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA; Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Rebecca C Schugar
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Amy Burrows
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Anthony D Gromovsky
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jenna Betters
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA
| | - Chase Neumann
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jessica Sacks
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Stephanie Marshall
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA; Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Russell Watts
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Richard G Lee
- Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Rosanne M Crooke
- Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Mark J Graham
- Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Takuya F Sakaguchi
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Richard Lehner
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - J Mark Brown
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
38
|
Zhang W, Bu SY, Mashek MT, O-Sullivan I, Sibai Z, Khan SA, Ilkayeva O, Newgard CB, Mashek DG, Unterman TG. Integrated Regulation of Hepatic Lipid and Glucose Metabolism by Adipose Triacylglycerol Lipase and FoxO Proteins. Cell Rep 2016; 15:349-59. [PMID: 27050511 DOI: 10.1016/j.celrep.2016.03.021] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 02/23/2016] [Accepted: 03/03/2016] [Indexed: 12/16/2022] Open
Abstract
Metabolism is a highly integrated process that is coordinately regulated between tissues and within individual cells. FoxO proteins are major targets of insulin action and contribute to the regulation of gluconeogenesis, glycolysis, and lipogenesis in the liver. However, the mechanisms by which FoxO proteins exert these diverse effects in an integrated fashion remain poorly understood. We report that FoxO proteins also exert important effects on intrahepatic lipolysis and fatty acid oxidation via the regulation of adipose triacylglycerol lipase (ATGL), which mediates the first step in lipolysis, and its inhibitor, the G0/S1 switch 2 gene (G0S2). We also find that ATGL-dependent lipolysis plays a critical role in mediating diverse effects of FoxO proteins in the liver, including effects on gluconeogenic, glycolytic, and lipogenic gene expression and metabolism. These results indicate that intrahepatic lipolysis plays a critical role in mediating and integrating the regulation of glucose and lipid metabolism downstream of FoxO proteins.
Collapse
Affiliation(s)
- Wenwei Zhang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Medical Research Service, Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - So Young Bu
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mara T Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - InSug O-Sullivan
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Medical Research Service, Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Zakaria Sibai
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Medical Research Service, Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Salmaan A Khan
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC 27710, USA; Department of Pharmacology, Duke University, Durham, NC 27710, USA; Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC 27710, USA; Department of Pharmacology, Duke University, Durham, NC 27710, USA; Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Terry G Unterman
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Medical Research Service, Jesse Brown VA Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
39
|
Doler C, Schweiger M, Zimmermann R, Breinbauer R. Chemical Genetic Approaches for the Investigation of Neutral Lipid Metabolism. Chembiochem 2016; 17:358-77. [DOI: 10.1002/cbic.201500501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Carina Doler
- Institute of Organic Chemistry; Graz University of Technology; Stremayrgasse 9 8010 Graz Austria
| | - Martina Schweiger
- Institute of Molecular Biosciences; University of Graz; Heinrichstrasse 31/II 8010 Graz Austria
| | - Robert Zimmermann
- Institute of Molecular Biosciences; University of Graz; Heinrichstrasse 31/II 8010 Graz Austria
| | - Rolf Breinbauer
- Institute of Organic Chemistry; Graz University of Technology; Stremayrgasse 9 8010 Graz Austria
| |
Collapse
|
40
|
Effect of Bariatric Weight Loss on the Adipose Lipolytic Transcriptome in Obese Humans. Mediators Inflamm 2015; 2015:106237. [PMID: 26663986 PMCID: PMC4667060 DOI: 10.1155/2015/106237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/02/2015] [Indexed: 01/04/2023] Open
Abstract
Background. Dysregulated lipolysis has been implicated in mechanisms of cardiometabolic disease and inflammation in obesity. Purpose. We sought to examine the effect of bariatric weight loss on adipose tissue lipolytic gene expression and their relationship to systemic metabolic parameters in obese subjects. Methods/Results. We biopsied subcutaneous adipose tissue in 19 obese individuals (BMI 42 ± 5 kg/m2, 79% female) at baseline and after a mean period of 8 ± 5 months (range 3–15 months) following bariatric surgery. We performed adipose tissue mRNA expression of proteins involved in triglyceride hydrolysis and correlated their weight loss induced alterations with systemic parameters associated with cardiovascular disease risk. mRNA transcripts of adipose triglyceride lipase (ATGL), hormone-sensitive lipase (HSL), and lipid droplet proteins comparative gene identification 58 (CGI-58) and perilipin increased significantly after weight loss (p < 0.05 for all). ATGL expression correlated inversely with plasma triglyceride (TG), hemoglobin A1C (HbA1C), and glucose, and HSL expression correlated negatively with glucose, while CGI-58 was inversely associated with HbA1C. Conclusion. We observed increased expression of adipose tissue lipolytic genes following bariatric weight loss which correlated inversely with systemic markers of lipid and glucose metabolism. Functional alterations in lipolysis in human adipose tissue may play a role in shaping cardiometabolic phenotypes in human obesity.
Collapse
|
41
|
Morigny P, Houssier M, Mouisel E, Langin D. Adipocyte lipolysis and insulin resistance. Biochimie 2015; 125:259-66. [PMID: 26542285 DOI: 10.1016/j.biochi.2015.10.024] [Citation(s) in RCA: 316] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 10/30/2015] [Indexed: 12/15/2022]
Abstract
Obesity-induced insulin resistance is a major risk factor for the development of type 2 diabetes. Basal fat cell lipolysis (i.e., fat cell triacylglycerol breakdown into fatty acids and glycerol in the absence of stimulatory factors) is elevated during obesity and is closely associated with insulin resistance. Inhibition of adipocyte lipolysis may therefore be a promising therapeutic strategy for treating insulin resistance and preventing obesity-associated type 2 diabetes. In this review, we explore the relationship between adipose lipolysis and insulin sensitivity. After providing an overview of the components of fat cell lipolytic machinery, we describe the hypotheses that may support the causality between lipolysis and insulin resistance. Excessive circulating fatty acids may ectopically accumulate in insulin-sensitive tissues and impair insulin action. Increased basal lipolysis may also modify the secretory profile of adipose tissue, influencing whole body insulin sensitivity. Finally, excessive fatty acid release may also worsen adipose tissue inflammation, a well-known parameter contributing to insulin resistance. Partial genetic or pharmacologic inhibition of fat cell lipases in mice as well as short term clinical trials using antilipolytic drugs in humans support the benefit of fat cell lipolysis inhibition on systemic insulin sensitivity and glucose metabolism, which occurs without an increase of fat mass. Modulation of fatty acid fluxes and, putatively, of fat cell secretory pattern may explain the amelioration of insulin sensitivity whereas changes in adipose tissue immune response do not seem involved.
Collapse
Affiliation(s)
- Pauline Morigny
- INSERM, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, UMR1048, Paul Sabatier University, France
| | - Marianne Houssier
- INSERM, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, UMR1048, Paul Sabatier University, France
| | - Etienne Mouisel
- INSERM, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, UMR1048, Paul Sabatier University, France
| | - Dominique Langin
- INSERM, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, UMR1048, Paul Sabatier University, France; Toulouse University Hospitals, Department of Clinical Biochemistry, Toulouse, France.
| |
Collapse
|
42
|
Hypophagia and metabolic adaptations in mice with defective ATGL-mediated lipolysis cause resistance to HFD-induced obesity. Proc Natl Acad Sci U S A 2015; 112:13850-5. [PMID: 26508640 DOI: 10.1073/pnas.1516004112] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Adipose triglyceride lipase (ATGL) initiates intracellular triglyceride (TG) catabolism. In humans, ATGL deficiency causes neutral lipid storage disease with myopathy (NLSDM) characterized by a systemic TG accumulation. Mice with a genetic deletion of ATGL (AKO) also accumulate TG in many tissues. However, neither NLSDM patients nor AKO mice are exceedingly obese. This phenotype is unexpected considering the importance of the enzyme for TG catabolism in white adipose tissue (WAT). In this study, we identified the counteracting mechanisms that prevent excessive obesity in the absence of ATGL. We used "healthy" AKO mice expressing ATGL exclusively in cardiomyocytes (AKO/cTg) to circumvent the cardiomyopathy and premature lethality observed in AKO mice. AKO/cTg mice were protected from high-fat diet (HFD)-induced obesity despite complete ATGL deficiency in WAT and normal adipocyte differentiation. AKO/cTg mice were highly insulin sensitive under hyperinsulinemic-euglycemic clamp conditions, eliminating insulin insensitivity as a possible protective mechanism. Instead, reduced food intake and altered signaling by peroxisome proliferator-activated receptor-gamma (PPAR-γ) and sterol regulatory element binding protein-1c in WAT accounted for the phenotype. These adaptations led to reduced lipid synthesis and storage in WAT of HFD-fed AKO/cTg mice. Treatment with the PPAR-γ agonist rosiglitazone reversed the phenotype. These results argue for the existence of an adaptive interdependence between lipolysis and lipid synthesis. Pharmacological inhibition of ATGL may prove useful to prevent HFD-induced obesity and insulin resistance.
Collapse
|
43
|
Schoiswohl G, Stefanovic-Racic M, Menke MN, Wills RC, Surlow BA, Basantani MK, Sitnick MT, Cai L, Yazbeck CF, Stolz DB, Pulinilkunnil T, O'Doherty RM, Kershaw EE. Impact of Reduced ATGL-Mediated Adipocyte Lipolysis on Obesity-Associated Insulin Resistance and Inflammation in Male Mice. Endocrinology 2015; 156. [PMID: 26196542 PMCID: PMC4588821 DOI: 10.1210/en.2015-1322] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Emerging evidence suggests that impaired regulation of adipocyte lipolysis contributes to the proinflammatory immune cell infiltration of metabolic tissues in obesity, a process that is proposed to contribute to the development and exacerbation of insulin resistance. To test this hypothesis in vivo, we generated mice with adipocyte-specific deletion of adipose triglyceride lipase (ATGL), the rate-limiting enzyme catalyzing triacylglycerol hydrolysis. In contrast to previous models, adiponectin-driven Cre expression was used for targeted ATGL deletion. The resulting adipocyte-specific ATGL knockout (AAKO) mice were then characterized for metabolic and immune phenotypes. Lean and diet-induced obese AAKO mice had reduced adipocyte lipolysis, serum lipids, systemic lipid oxidation, and expression of peroxisome proliferator-activated receptor alpha target genes in adipose tissue (AT) and liver. These changes did not increase overall body weight or fat mass in AAKO mice by 24 weeks of age, in part due to reduced expression of genes involved in lipid uptake, synthesis, and adipogenesis. Systemic glucose and insulin tolerance were improved in AAKO mice, primarily due to enhanced hepatic insulin signaling, which was accompanied by marked reduction in diet-induced hepatic steatosis as well as hepatic immune cell infiltration and activation. In contrast, although adipocyte ATGL deletion reduced AT immune cell infiltration in response to an acute lipolytic stimulus, it was not sufficient to ameliorate, and may even exacerbate, chronic inflammatory changes that occur in AT in response to diet-induced obesity.
Collapse
Affiliation(s)
- Gabriele Schoiswohl
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Maja Stefanovic-Racic
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Marie N Menke
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Rachel C Wills
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Beth A Surlow
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Mahesh K Basantani
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Mitch T Sitnick
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Lingzhi Cai
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Cynthia F Yazbeck
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Donna B Stolz
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Thomas Pulinilkunnil
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Robert M O'Doherty
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Erin E Kershaw
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| |
Collapse
|
44
|
Impaired mitochondrial fat oxidation induces adaptive remodeling of muscle metabolism. Proc Natl Acad Sci U S A 2015; 112:E3300-9. [PMID: 26056297 DOI: 10.1073/pnas.1418560112] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The correlations between intramyocellular lipid (IMCL), decreased fatty acid oxidation (FAO), and insulin resistance have led to the hypothesis that impaired FAO causes accumulation of lipotoxic intermediates that inhibit muscle insulin signaling. Using a skeletal muscle-specific carnitine palmitoyltransferase-1 KO model, we show that prolonged and severe mitochondrial FAO inhibition results in increased carbohydrate utilization, along with reduced physical activity; increased circulating nonesterified fatty acids; and increased IMCLs, diacylglycerols, and ceramides. Perhaps more importantly, inhibition of mitochondrial FAO also initiates a local, adaptive response in muscle that invokes mitochondrial biogenesis, compensatory peroxisomal fat oxidation, and amino acid catabolism. Loss of its major fuel source (lipid) induces an energy deprivation response in muscle coordinated by signaling through AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) to maintain energy supply for locomotion and survival. At the whole-body level, these adaptations result in resistance to obesity.
Collapse
|
45
|
Ramanadham S, Ali T, Ashley JW, Bone RN, Hancock WD, Lei X. Calcium-independent phospholipases A2 and their roles in biological processes and diseases. J Lipid Res 2015; 56:1643-68. [PMID: 26023050 DOI: 10.1194/jlr.r058701] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Indexed: 12/24/2022] Open
Abstract
Among the family of phospholipases A2 (PLA2s) are the Ca(2+)-independent PLA2s (iPLA2s) and they are designated group VI iPLA2s. In relation to secretory and cytosolic PLA2s, the iPLA2s are more recently described and details of their expression and roles in biological functions are rapidly emerging. The iPLA2s or patatin-like phospholipases (PNPLAs) are intracellular enzymes that do not require Ca(2+) for activity, and contain lipase (GXSXG) and nucleotide-binding (GXGXXG) consensus sequences. Though nine PNPLAs have been recognized, PNPLA8 (membrane-associated iPLA2γ) and PNPLA9 (cytosol-associated iPLA2β) are the most widely studied and understood. The iPLA2s manifest a variety of activities in addition to phospholipase, are ubiquitously expressed, and participate in a multitude of biological processes, including fat catabolism, cell differentiation, maintenance of mitochondrial integrity, phospholipid remodeling, cell proliferation, signal transduction, and cell death. As might be expected, increased or decreased expression of iPLA2s can have profound effects on the metabolic state, CNS function, cardiovascular performance, and cell survival; therefore, dysregulation of iPLA2s can be a critical factor in the development of many diseases. This review is aimed at providing a general framework of the current understanding of the iPLA2s and discussion of the potential mechanisms of action of the iPLA2s and related involved lipid mediators.
Collapse
Affiliation(s)
- Sasanka Ramanadham
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Tomader Ali
- Undergraduate Research Office, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jason W Ashley
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104
| | - Robert N Bone
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - William D Hancock
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Xiaoyong Lei
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
46
|
Mayer N, Schweiger M, Melcher MC, Fledelius C, Zechner R, Zimmermann R, Breinbauer R. Structure-activity studies in the development of a hydrazone based inhibitor of adipose-triglyceride lipase (ATGL). Bioorg Med Chem 2015; 23:2904-16. [PMID: 25778769 PMCID: PMC4457358 DOI: 10.1016/j.bmc.2015.02.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 02/24/2015] [Accepted: 02/25/2015] [Indexed: 11/07/2022]
Abstract
Adipose triglyceride lipase (ATGL) catalyzes the degradation of cellular triacylglycerol stores and strongly determines the concentration of circulating fatty acids (FAs). High serum FA levels are causally linked to the development of insulin resistance and impaired glucose tolerance, which eventually progresses to overt type 2 diabetes. ATGL-specific inhibitors could be used to lower circulating FAs, which can counteract the development of insulin resistance. In this article, we report about structure–activity relationship (SAR) studies of small molecule inhibitors of ATGL based on a hydrazone chemotype. The SAR indicated that the binding pocket of ATGL requests rather linear compounds without bulky substituents. The best inhibitor showed an IC50 = 10 μM in an assay with COS7-cell lysate overexpressing murine ATGL.
Collapse
Affiliation(s)
- Nicole Mayer
- Institute of Organic Chemistry, Graz University of Technology, Stremayrgasse 9, A-8010 Graz, Austria
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, Heinrichstraße 31/II, A-8010 Graz, Austria
| | | | | | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Heinrichstraße 31/II, A-8010 Graz, Austria
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Heinrichstraße 31/II, A-8010 Graz, Austria.
| | - Rolf Breinbauer
- Institute of Organic Chemistry, Graz University of Technology, Stremayrgasse 9, A-8010 Graz, Austria.
| |
Collapse
|
47
|
Coelho M, Nunes P, Mendes VM, Manadas B, Heerschap A, Jones JG. Effect of Global ATGL Knockout on Murine Fasting Glucose Kinetics. J Diabetes Res 2015; 2015:542029. [PMID: 26236747 PMCID: PMC4506825 DOI: 10.1155/2015/542029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/08/2015] [Indexed: 12/25/2022] Open
Abstract
Mice deficient in adipose triglyceride lipase (ATGL(-/-)) present elevated ectopic lipid levels but are paradoxically glucose-tolerant. Measurement of endogenous glucose production (EGP) and Cori cycle activity provide insights into the maintenance of glycemic control in these animals. These parameters were determined in 7 wild-type (ATGL(+/-)) and 6 ATGL(-/-) mice by a primed-infusion of [U-(13)C6]glucose followed by LC-MS/MS targeted mass-isotopomer analysis of blood glucose. EGP was quantified by isotope dilution of [U-(13)C6]glucose while Cori cycling was estimated by analysis of glucose triose (13)C-isotopomers. Fasting plasma free fatty-acids were significantly lower in ATGL(-/-) versus control mice (0.43 ± 0.05 mM versus 0.73 ± 0.11 mM, P < 0.05). Six-hour fasting EGP rates were identical for both ATGL(-/-) and control mice (79 ± 11 versus 71 ± 7 μmol/kg/min, resp.). Peripheral glucose metabolism was dominated by Cori cycling (80 ± 2% and 82 ± 7% of glucose disposal for ATGL(-/-) and control mice, resp.) indicating that peripheral glucose oxidation was not significantly upregulated in ATGL(-/-) mice under these conditions. The glucose (13)C-isotopomer distributions in both ATGL(-/-) and control mice were consistent with extensive hepatic pyruvate recycling. This suggests that gluconeogenic outflow from the Krebs cycle was also well compensated in ATGL(-/-) mice.
Collapse
Affiliation(s)
- Margarida Coelho
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | | | - Vera M. Mendes
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Bruno Manadas
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Arend Heerschap
- Department of Radiology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - John G. Jones
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Portuguese Diabetes Association (APDP), Lisbon, Portugal
- *John G. Jones:
| |
Collapse
|
48
|
Heier C, Zimmermann R. The sparing use of fat: G0s2 controls lipolysis and fatty acid oxidation. Diabetologia 2015; 58:7-9. [PMID: 25351607 DOI: 10.1007/s00125-014-3430-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 10/06/2014] [Indexed: 10/24/2022]
Affiliation(s)
- Christoph Heier
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31A, 8010, Graz, Austria
| | | |
Collapse
|
49
|
Gauthier MS, Pérusse JR, Lavoie MÈ, Sladek R, Madiraju SRM, Ruderman NB, Coulombe B, Prentki M, Rabasa-Lhoret R. Increased subcutaneous adipose tissue expression of genes involved in glycerolipid-fatty acid cycling in obese insulin-resistant versus -sensitive individuals. J Clin Endocrinol Metab 2014; 99:E2518-28. [PMID: 25210878 PMCID: PMC5393488 DOI: 10.1210/jc.2014-1662] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT A subpopulation of obese individuals remains insulin sensitive (ISO). They represent a unique human model to investigate factors underlying insulin resistance (IR) without the confounding effect of major differences in weight/adiposity. Altered fatty-acid (FA) metabolism in sc adipose tissue (SAT) contributes to obesity-associated IR. OBJECTIVE To test the hypothesis that ISO and body mass index-matched insulin-resistant obese (IRO) patients demonstrate differential SAT expression profiles of genes involved in glycerolipid-FA metabolism and that weight loss-induced improvement of IR ameliorates these changes. DESIGN AND SETTING A cross-sectional and longitudinal study. PATIENTS AND INTERVENTION Thirty-eight nondiabetic obese women were stratified into ISO (n = 25) or IRO (n = 13) groups based on hyperinsulinemic-euglycemic clamp results. Subjects were studied before and after a 6-month hypocaloric diet intervention. MAIN OUTCOME MEASURES mRNA (quantitative RT-PCR) and protein (mass spectrometry and immunoblots) levels were measured in SAT biopsies. RESULTS Despite having age, body mass index, and fat mass similar to ISO individuals, IRO patients had lower insulin sensitivity and glucose tolerance (P < .05). Baseline SAT mRNA and protein levels of genes involved in both the synthesis and lipolysis of glycerolipid-FAs were higher in IRO individuals (P < .05), even when groups were matched for visceral adipose tissue content. The dietary intervention resulted in approximately 6% weight loss in both the IRO and ISO groups (P < .05) but only ameliorated insulin sensitivity in IRO individuals (P < .05). Likewise, the intervention reduced the expression of most glycerolipid-FA metabolism genes (P < .05), with expression levels in IRO individuals being restored to ISO levels. CONCLUSIONS Increased SAT expression of genes involved in both the synthesis and hydrolysis of glycerolipid-FAs is closely associated with IR in obese women. The results suggest that enhanced glycerolipid-FA cycling in SAT contributes to obesity-associated IR.
Collapse
Affiliation(s)
- Marie-Soleil Gauthier
- Institut de recherches cliniques de Montréal (M.-S.G., J.R.P., M.-E.L., B.C., R.R.-L.), Montréal, QC H2W 1R7, Canada; Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) (M.-S.G., M.-E.L., R.S., S.R.M.M., M.P., R.R.-L.), Montréal, QC H2X 0A9, Canada; McGill University and Centre d'Innovation Génome Québec (R.S.), Montréal, QC H3A 0G1, Canada; Molecular Nutrition Unit at the CRCHUM (S.R.M.M., M.P.), Montréal, QC H2X 0A9, Canada; Diabetes and Metabolism Research Unit (N.B.R.), and Department of Medicine and Section of Endocrinology (N.B.R.), Boston University School of Medicine, Boston, Massachusetts 02118; Departments of Biochemistry (B.C., M.P.) and Nutrition (M.-E.L., M.P., R.R.-L.), Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Catecholamine-induced lipolysis causes mTOR complex dissociation and inhibits glucose uptake in adipocytes. Proc Natl Acad Sci U S A 2014; 111:17450-5. [PMID: 25422441 DOI: 10.1073/pnas.1410530111] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Anabolic and catabolic signaling oppose one another in adipose tissue to maintain cellular and organismal homeostasis, but these pathways are often dysregulated in metabolic disorders. Although it has long been established that stimulation of the β-adrenergic receptor inhibits insulin-stimulated glucose uptake in adipocytes, the mechanism has remained unclear. Here we report that β-adrenergic-mediated inhibition of glucose uptake requires lipolysis. We also show that lipolysis suppresses glucose uptake by inhibiting the mammalian target of rapamycin (mTOR) complexes 1 and 2 through complex dissociation. In addition, we show that products of lipolysis inhibit mTOR through complex dissociation in vitro. These findings reveal a previously unrecognized intracellular signaling mechanism whereby lipolysis blocks the phosphoinositide 3-kinase-Akt-mTOR pathway, resulting in decreased glucose uptake. This previously unidentified mechanism of mTOR regulation likely contributes to the development of insulin resistance.
Collapse
|