201
|
Peroxisome Proliferator-Activated Receptor γ and Its Role in Adipocyte Homeostasis and Thiazolidinedione-Mediated Insulin Sensitization. Mol Cell Biol 2018; 38:MCB.00677-17. [PMID: 29483301 DOI: 10.1128/mcb.00677-17] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 02/20/2018] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue is a dynamic organ that makes critical contributions to whole-body metabolic homeostasis. Although recent studies have revealed that different fat depots have distinct molecular signatures, metabolic functions and adipogenic mechanisms, peroxisome proliferator-activated receptor γ (PPARγ) is still widely viewed as the master regulator of adipogenesis and critical for maintaining mature adipocyte function. Using an inducible, adipocyte-specific knockout system, we explored the role of PPARγ in mature adipocytes in vivo Short-term PPARγ deficiency in adipocytes reduces whole-body insulin sensitivity, but adipocytes are viable both in vitro and in vivo However, after exposure to a high-fat diet, even short-term PPARγ deficiency leads to rapid adipocyte death. When mature adipocytes are depleted of both PPARγ and CCAAT-enhancer-binding protein α (C/EBPα), they are rapidly depleted of lipids and undergo adipocyte death, both in vitro and in vivo Surprisingly, although thiazolidinediones (TZDs; PPARγ agonists) are thought to act mainly on PPARγ, PPARγ in adipocytes is not required for the whole-body insulin-sensitizing effect of TZDs. This offers new mechanistic aspects of PPARγ/TZD action and its effect on whole-body metabolic homeostasis.
Collapse
|
202
|
Obata Y, Kita S, Koyama Y, Fukuda S, Takeda H, Takahashi M, Fujishima Y, Nagao H, Masuda S, Tanaka Y, Nakamura Y, Nishizawa H, Funahashi T, Ranscht B, Izumi Y, Bamba T, Fukusaki E, Hanayama R, Shimada S, Maeda N, Shimomura I. Adiponectin/T-cadherin system enhances exosome biogenesis and decreases cellular ceramides by exosomal release. JCI Insight 2018; 3:99680. [PMID: 29669945 DOI: 10.1172/jci.insight.99680] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/14/2018] [Indexed: 12/15/2022] Open
Abstract
Adiponectin, an adipocyte-derived circulating protein, accumulates in vasculature, heart, and skeletal muscles through interaction with a unique glycosylphosphatidylinositol-anchored cadherin, T-cadherin. Recent studies have demonstrated that such accumulation is essential for adiponectin-mediated cardiovascular protection. Here, we demonstrate that the adiponectin/T-cadherin system enhances exosome biogenesis and secretion, leading to the decrease of cellular ceramides. Adiponectin accumulated inside multivesicular bodies, the site of exosome generation, in cultured cells and in vivo aorta, and also in exosomes in conditioned media and in blood, together with T-cadherin. The systemic level of exosomes in blood was significantly affected by adiponectin or T-cadherin in vivo. Adiponectin increased exosome biogenesis from the cells, dependently on T-cadherin, but not on AdipoR1 or AdipoR2. Such enhancement of exosome release accompanied the reduction of cellular ceramides through ceramide efflux in exosomes. Consistently, the ceramide reduction by adiponectin was found in aortas of WT mice treated with angiotensin II, but not in T-cadherin-knockout mice. Our findings provide insights into adiponectin/T-cadherin-mediated organ protection through exosome biogenesis and secretion.
Collapse
Affiliation(s)
| | - Shunbun Kita
- Department of Metabolic Medicine.,Department of Adipose Management, and
| | - Yoshihisa Koyama
- Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | - Hiroaki Takeda
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | - Tohru Funahashi
- Department of Metabolic Medicine.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Barbara Ranscht
- Sanford Burnham Prebys Medical Discovery Institute, NIH-designated Cancer Center, Development, Aging and Regeneration Program, La Jolla, California, USA
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Eiichiro Fukusaki
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Rikinari Hanayama
- Department of Immunology, Kanazawa University Graduate School of Medical Sciences, Ishikawa, Japan
| | - Shoichi Shimada
- Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Norikazu Maeda
- Department of Metabolic Medicine.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | |
Collapse
|
203
|
Hernandez-Carretero A, Weber N, La Frano MR, Ying W, Rodriguez JL, Sears DD, Wallenius V, Börgeson E, Newman JW, Osborn O. Obesity-induced changes in lipid mediators persist after weight loss. Int J Obes (Lond) 2018; 42:728-736. [PMID: 29089614 PMCID: PMC6055936 DOI: 10.1038/ijo.2017.266] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 09/01/2017] [Accepted: 10/16/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Obesity induces significant changes in lipid mediators, however, the extent to which these changes persist after weight loss has not been investigated. SUBJECTS/METHODS We fed C57BL6 mice a high-fat diet to generate obesity and then switched the diet to a lower-fat diet to induce weight loss. We performed a comprehensive metabolic profiling of lipid mediators including oxylipins, endocannabinoids, sphingosines and ceramides in key metabolic tissues (including adipose, liver, muscle and hypothalamus) and plasma. RESULTS We found that changes induced by obesity were largely reversible in most metabolic tissues but the adipose tissue retained a persistent obese metabolic signature. Prostaglandin signaling was perturbed in the obese state and lasting increases in PGD2, and downstream metabolites 15-deoxy PGJ2 and delta-12-PGJ2 were observed after weight loss. Furthermore expression of the enzyme responsible for PGD2 synthesis (hematopoietic prostaglandin D synthase, HPGDS) was increased in obese adipose tissues and remained high after weight loss. We found that inhibition of HPGDS over the course of 5 days resulted in decreased food intake in mice. Increased HPGDS expression was also observed in human adipose tissues obtained from obese compared with lean individuals. We then measured circulating levels of PGD2 in obese patients before and after weight loss and found that while elevated relative to lean subjects, levels of this metabolite did not decrease after significant weight loss. CONCLUSIONS These results suggest that lasting changes in lipid mediators induced by obesity, still present after weight loss, may play a role in the biological drive to regain weight.
Collapse
Affiliation(s)
| | - Natalie Weber
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, CA, USA
| | - Michael R. La Frano
- Department of Nutrition, University of California, Davis, CA, USA
- NIH West Coast Metabolomics Center, Davis, CA, USA
- Department of Food Science and Nutrition, California Polytechnic State University, San Luis Obispo, USA
| | - Wei Ying
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, CA, USA
| | - Juan Lantero Rodriguez
- The Wallenberg Laboratory for Cardiovascular and Metabolic Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Dorothy D. Sears
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, CA, USA
| | - Ville Wallenius
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Emma Börgeson
- The Wallenberg Laboratory for Cardiovascular and Metabolic Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - John W. Newman
- Department of Nutrition, University of California, Davis, CA, USA
- NIH West Coast Metabolomics Center, Davis, CA, USA
- Obesity and Metabolism Research Unit, USDA-ARS-Western Human Nutrition Research Center, Davis, CA, USA
| | - Olivia Osborn
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, CA, USA
| |
Collapse
|
204
|
Xia JY, Sun K, Hepler C, Ghaben AL, Gupta RK, An YA, Holland WL, Morley TS, Adams AC, Gordillo R, Kusminski CM, Scherer PE. Acute loss of adipose tissue-derived adiponectin triggers immediate metabolic deterioration in mice. Diabetologia 2018; 61:932-941. [PMID: 29224189 PMCID: PMC5844860 DOI: 10.1007/s00125-017-4516-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/03/2017] [Indexed: 01/08/2023]
Abstract
AIM/HYPOTHESIS Adiponectin (APN), a circulating hormone secreted by mature adipocytes, has been extensively studied because it has beneficial metabolic effects. While many studies have focused on the congenital loss of APN and its effects on systemic body glucose and lipid metabolism, little is known about the effects triggered by acute loss of APN in the adult mouse. We anticipated that genetically induced acute depletion of APN in adult mice would have a more profound effect on systemic metabolic health than congenital deletion of Adipoq, the gene encoding APN, with its associated potential for adaptive responses that may mask the phenotypes. METHODS Mice carrying loxP-flanked regions of Adipoq were generated and bred to the Adipoq (APN) promoter-driven reverse tetracycline-controlled transactivator (rtTA) (APN-rtTA) gene and a tet-responsive Cre line (TRE-Cre) to achieve acute depletion of APN. Upon acute removal of APN in adult mice, systemic glucose and lipid homeostasis were assessed under basal and insulinopenic conditions. RESULTS The acute depletion of APN results in more severe systemic insulin resistance and hyperlipidaemia than in mice with congenital loss of APN. Furthermore, the acute depletion of APN in adult mice results in a much more dramatic reduction in survival rate, with 50% of inducible knockouts dying in the first 5 days under insulinopenic conditions compared with 0% of congenital Adipoq knockout mice under similar conditions. CONCLUSIONS/INTERPRETATION Acute systemic removal of APN results in a much more negative metabolic phenotype compared with congenital knockout of Adipoq. Specifically, our data demonstrate that acute depletion of APN is especially detrimental to lipid homeostasis, both under basal and insulinopenic conditions. This suggests that compensatory mechanisms exist in congenital knockout mice that offset some of the metabolic actions covered by APN.
Collapse
Affiliation(s)
- Jonathan Y Xia
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
- McGaw Medical Center of Northwestern University, Department of Internal Medicine, Chicago, IL, USA
| | - Kai Sun
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chelsea Hepler
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - Alexandra L Ghaben
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - Yu A An
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - William L Holland
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - Thomas S Morley
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - Andrew C Adams
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC8549, Dallas, TX, 75390-8549, USA.
- Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
205
|
Aryal B, Singh AK, Zhang X, Varela L, Rotllan N, Goedeke L, Chaube B, Camporez JP, Vatner DF, Horvath TL, Shulman GI, Suárez Y, Fernández-Hernando C. Absence of ANGPTL4 in adipose tissue improves glucose tolerance and attenuates atherogenesis. JCI Insight 2018; 3:97918. [PMID: 29563332 PMCID: PMC5926923 DOI: 10.1172/jci.insight.97918] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 02/14/2018] [Indexed: 12/12/2022] Open
Abstract
Alterations in ectopic lipid deposition and circulating lipids are major risk factors for developing cardiometabolic diseases. Angiopoietin-like protein 4 (ANGPTL4), a protein that inhibits lipoprotein lipase (LPL), controls fatty acid (FA) uptake in adipose and oxidative tissues and regulates circulating triacylglycerol-rich (TAG-rich) lipoproteins. Unfortunately, global depletion of ANGPTL4 results in severe metabolic abnormalities, inflammation, and fibrosis when mice are fed a high-fat diet (HFD), limiting our understanding of the contribution of ANGPTL4 in metabolic disorders. Here, we demonstrate that genetic ablation of ANGPTL4 in adipose tissue (AT) results in enhanced LPL activity, rapid clearance of circulating TAGs, increased AT lipolysis and FA oxidation, and decreased FA synthesis in AT. Most importantly, we found that absence of ANGPTL4 in AT prevents excessive ectopic lipid deposition in the liver and muscle, reducing novel PKC (nPKC) membrane translocation and enhancing insulin signaling. As a result, we observed a remarkable improvement in glucose tolerance in short-term HFD-fed AT-specific Angptl4-KO mice. Finally, lack of ANGPTL4 in AT enhances the clearance of proatherogenic lipoproteins, attenuates inflammation, and reduces atherosclerosis. Together, these findings uncovered an essential role of AT ANGPTL4 in regulating peripheral lipid deposition, influencing whole-body lipid and glucose metabolism and the progression of atherosclerosis.
Collapse
Affiliation(s)
- Binod Aryal
- Vascular Biology and Therapeutics Program
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
| | - Abhishek K. Singh
- Vascular Biology and Therapeutics Program
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
| | - Xinbo Zhang
- Vascular Biology and Therapeutics Program
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
| | - Luis Varela
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
| | - Noemi Rotllan
- Vascular Biology and Therapeutics Program
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
| | | | - Balkrishna Chaube
- Vascular Biology and Therapeutics Program
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
| | | | | | - Tamas L. Horvath
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
| | - Gerald I. Shulman
- Department of Internal Medicine
- Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, and
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
206
|
Ter Horst KW, Gilijamse PW, Versteeg RI, Ackermans MT, Nederveen AJ, la Fleur SE, Romijn JA, Nieuwdorp M, Zhang D, Samuel VT, Vatner DF, Petersen KF, Shulman GI, Serlie MJ. Hepatic Diacylglycerol-Associated Protein Kinase Cε Translocation Links Hepatic Steatosis to Hepatic Insulin Resistance in Humans. Cell Rep 2018; 19:1997-2004. [PMID: 28591572 PMCID: PMC5469939 DOI: 10.1016/j.celrep.2017.05.035] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/17/2017] [Accepted: 05/10/2017] [Indexed: 12/12/2022] Open
Abstract
Hepatic lipid accumulation has been implicated in the development of insulin resistance, but translational evidence in humans is limited. We investigated the relationship between liver fat and tissue-specific insulin sensitivity in 133 obese subjects. Although the presence of hepatic steatosis in obese subjects was associated with hepatic, adipose tissue, and peripheral insulin resistance, we found that intrahepatic triglycerides were not strictly sufficient or essential for hepatic insulin resistance. Thus, to examine the molecular mechanisms that link hepatic steatosis to hepatic insulin resistance, we comprehensively analyzed liver biopsies from a subset of 29 subjects. Here, hepatic cytosolic diacylglycerol content, but not hepatic ceramide content, was increased in subjects with hepatic insulin resistance. Moreover, cytosolic diacylglycerols were strongly associated with hepatic PKCε activation, as reflected by PKCε translocation to the plasma membrane. These results demonstrate the relevance of hepatic diacylglycerol-induced PKCε activation in the pathogenesis of NAFLD-associated hepatic insulin resistance in humans. The presence of hepatic steatosis is associated with insulin resistance Intrahepatic triglycerides are not sufficient for hepatic insulin resistance Diacylglycerol in hepatic cytosol predicts insulin inhibition of glucose production Diacylglycerol-associated insulin resistance is characterized by PKCε translocation
Collapse
Affiliation(s)
- Kasper W Ter Horst
- Department of Endocrinology and Metabolism, Academic Medical Center, 1105AZ Amsterdam, the Netherlands
| | - Pim W Gilijamse
- Department of Endocrinology and Metabolism, Academic Medical Center, 1105AZ Amsterdam, the Netherlands
| | - Ruth I Versteeg
- Department of Endocrinology and Metabolism, Academic Medical Center, 1105AZ Amsterdam, the Netherlands
| | - Mariette T Ackermans
- Department of Clinical Chemistry, Laboratory of Endocrinology, Academic Medical Center, 1105AZ Amsterdam, the Netherlands
| | - Aart J Nederveen
- Department of Radiology, Academic Medical Center, 1105AZ Amsterdam, the Netherlands
| | - Susanne E la Fleur
- Department of Endocrinology and Metabolism, Academic Medical Center, 1105AZ Amsterdam, the Netherlands; Department of Clinical Chemistry, Laboratory of Endocrinology, Academic Medical Center, 1105AZ Amsterdam, the Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, 1105BA Amsterdam, the Netherlands
| | - Johannes A Romijn
- Department of Medicine, Academic Medical Center, 1105AZ Amsterdam, the Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Academic Medical Center, 1105AZ Amsterdam, the Netherlands; Department of Internal Medicine, VU University Medical Center, 1081HV Amsterdam, the Netherlands; Institute for Cardiovascular Research, VU University Medical Center, 1081HV Amsterdam, the Netherlands
| | - Dongyan Zhang
- Howard Hughes Medical Institute, Yale University, New Haven, CT 06519, USA
| | - Varman T Samuel
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Daniel F Vatner
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Kitt F Petersen
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gerald I Shulman
- Howard Hughes Medical Institute, Yale University, New Haven, CT 06519, USA; Department of Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Academic Medical Center, 1105AZ Amsterdam, the Netherlands.
| |
Collapse
|
207
|
Abstract
Studies of bioactive lipids in general and sphingolipids in particular have intensified over the past several years, revealing an unprecedented and unanticipated complexity of the lipidome and its many functions, which rivals, if not exceeds, that of the genome or proteome. These results highlight critical roles for bioactive sphingolipids in most, if not all, major cell biological responses, including all major cell signalling pathways, and they link sphingolipid metabolism to key human diseases. Nevertheless, the fairly nascent field of bioactive sphingolipids still faces challenges in its biochemical and molecular underpinnings, including defining the molecular mechanisms of pathway and enzyme regulation, the study of lipid-protein interactions and the development of cellular probes, suitable biomarkers and therapeutic approaches.
Collapse
Affiliation(s)
- Yusuf A Hannun
- Stony Brook Cancer Center and Department of Medicine, Stony Brook University, New York 11794, USA
| | - Lina M Obeid
- Stony Brook Cancer Center and Department of Medicine, Stony Brook University, New York 11794, USA
- Northport Veterans Affairs Medical Center, Northport, New York 11768, USA
| |
Collapse
|
208
|
Ho N, Xu C, Thibault G. From the unfolded protein response to metabolic diseases - lipids under the spotlight. J Cell Sci 2018; 131:131/3/jcs199307. [PMID: 29439157 DOI: 10.1242/jcs.199307] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The unfolded protein response (UPR) is classically viewed as a stress response pathway to maintain protein homeostasis at the endoplasmic reticulum (ER). However, it has recently emerged that the UPR can be directly activated by lipid perturbation, independently of misfolded proteins. Comprising primarily phospholipids, sphingolipids and sterols, individual membranes can contain hundreds of distinct lipids. Even with such complexity, lipid distribution in a cell is tightly regulated by mechanisms that remain incompletely understood. It is therefore unsurprising that lipid dysregulation can be a key factor in disease development. Recent advances in analysis of lipids and their regulators have revealed remarkable mechanisms and connections to other cellular pathways including the UPR. In this Review, we summarize the current understanding in UPR transducers functioning as lipid sensors and the interplay between lipid metabolism and ER homeostasis in the context of metabolic diseases. We attempt to provide a framework consisting of a few key principles to integrate the different lines of evidence and explain this rather complicated mechanism.
Collapse
Affiliation(s)
- Nurulain Ho
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551
| | - Chengchao Xu
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142-1479, USA
| | - Guillaume Thibault
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551
| |
Collapse
|
209
|
Summers SA. Could Ceramides Become the New Cholesterol? Cell Metab 2018; 27:276-280. [PMID: 29307517 DOI: 10.1016/j.cmet.2017.12.003] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/15/2017] [Accepted: 12/01/2017] [Indexed: 12/24/2022]
Abstract
The Mayo Clinic recently introduced a diagnostic test that quantifies plasma ceramides in order to identify patients at risk of major adverse cardiac events. By comparing recent discoveries about these biomarker ceramides with the exhaustive body of literature surrounding cholesterol, Summers aims to highlight important advances and critically needed areas of investigation on this exciting class of bioactive lipids.
Collapse
Affiliation(s)
- Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
210
|
Marra F, Svegliati-Baroni G. Lipotoxicity and the gut-liver axis in NASH pathogenesis. J Hepatol 2018; 68:280-295. [PMID: 29154964 DOI: 10.1016/j.jhep.2017.11.014] [Citation(s) in RCA: 541] [Impact Index Per Article: 90.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 11/05/2017] [Accepted: 11/09/2017] [Indexed: 12/12/2022]
Abstract
The pathogenesis of non-alcoholic fatty liver disease, particularly the mechanisms whereby a minority of patients develop a more severe phenotype characterised by hepatocellular damage, inflammation, and fibrosis is still incompletely understood. Herein, we discuss two pivotal aspects of the pathogenesis of NASH. We first analyse the initial mechanisms responsible for hepatocellular damage and inflammation, which derive from the toxic effects of excess lipids. Accumulating data indicate that the total amount of triglycerides stored in hepatocytes is not the major determinant of lipotoxicity, and that specific lipid classes act as damaging agents on liver cells. In particular, the role of free fatty acids such as palmitic acid, cholesterol, lysophosphatidylcholine and ceramides has recently emerged. These lipotoxic agents affect the cell behaviour via multiple mechanisms, including activation of signalling cascades and death receptors, endoplasmic reticulum stress, modification of mitochondrial function, and oxidative stress. In the second part of this review, the cellular and molecular players involved in the cross-talk between the gut and the liver are considered. These include modifications to the microbiota, which provide signals through the intestine and bacterial products, as well as hormones produced in the bowel that affect metabolism at different levels including the liver. Finally, the activation of nuclear receptors by bile acids is analysed.
Collapse
Affiliation(s)
- Fabio Marra
- Dipartimento di Medicina Sperimentale e Clinica and Centro di Ricerca Denothe, Università di Firenze, Italy.
| | - Gianluca Svegliati-Baroni
- Dipartimento Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy; Centro Interdipartimentale Obesità, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
211
|
Azuma MM, Balani P, Boisvert H, Gil M, Egashira K, Yamaguchi T, Hasturk H, Duncan M, Kawai T, Movila A. Endogenous acid ceramidase protects epithelial cells from Porphyromonas gingivalis-induced inflammation in vitro. Biochem Biophys Res Commun 2018; 495:2383-2389. [PMID: 29278706 PMCID: PMC5765770 DOI: 10.1016/j.bbrc.2017.12.137] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 12/22/2017] [Indexed: 01/08/2023]
Abstract
Ceramidases are a group of enzymes that degrade pro-inflammatory ceramide by cleaving a fatty acid to form anti-inflammatory sphingosine lipid. Thus far, acid, neutral and alkaline ceramidase isozymes have been described. However, the expression patterns of ceramidase isoforms as well as their role in periodontal disease pathogenesis remain unknown. In this study, expression patterns of ceramidase isoforms were quantified by real-time PCR and immunohistochemistry in gingival samples of patients with periodontitis and healthy subjects, as well as in EpiGingivalTM-3D culture and OBA-9 gingival epithelial cells both of which were stimulated with or without the presence of live Porphyromonas gingivalis (ATCC 33277 strain). A significantly lower level of acid ceramidase expression was detected in gingival tissues from periodontal patients compared to those from healthy subjects. In addition, acid-ceramidase expression in EpiGingival™ 3D culture and OBA-9 cells was suppressed by stimulation with P. gingivalis in vitro. No significant fluctuation was detected for neutral or alkaline ceramidases in either gingival samples or cell cultures. Next, to elucidate the role of acid ceramidase in P. gingivalis-induced inflammation in vitro, OBA-9 cells were transduced with adenoviral vector expressing the human acid ceramidase (Ad-ASAH1) gene or control adenoviral vector (Ad-control). In response to stimulation with P. gingivalis, ASAH1-over-expressing OBA-9 cells showed significantly lower mRNA expressions of caspase-3 as well as the percentage of Annexin V-positive cells, when compared with OBA-9 cells transduced with Ad-control vector. Furthermore, in response to stimulation with P. gingivalis, ASAH1-over-expressing OBA-9 cells produced less TNF-α, IL-6, and IL1β pro-inflammatory cytokines than observed in OBA-9 cells transduced with Ad-control vector. Collectively, our data show the novel discovery of anti-inflammatory and anti-apoptotic effects of acid ceramidase in host cells exposed to periodontal bacteria, and the attenuation of the expression of host-protective acid ceramidase in periodontal lesions.
Collapse
Affiliation(s)
| | - Pooja Balani
- The Forsyth Institute, Cambridge, MA, USA; Harvard University School of Dental Medicine, Boston, MA, USA
| | | | - Mindy Gil
- The Forsyth Institute, Cambridge, MA, USA; Harvard University School of Dental Medicine, Boston, MA, USA
| | - Kenji Egashira
- The Forsyth Institute, Cambridge, MA, USA; Lion Corporation, Research & Development Headquarter, Odawara, Kanagawa, Japan
| | - Tsuguno Yamaguchi
- The Forsyth Institute, Cambridge, MA, USA; Lion Corporation, Research & Development Headquarter, Odawara, Kanagawa, Japan
| | - Hatice Hasturk
- The Forsyth Institute, Cambridge, MA, USA; Harvard University School of Dental Medicine, Boston, MA, USA
| | | | - Toshihisa Kawai
- NOVA Southeastern University, College of Dental Medicine, Fort Lauderdale, FL, USA
| | - Alexandru Movila
- The Forsyth Institute, Cambridge, MA, USA; Harvard University School of Dental Medicine, Boston, MA, USA; NOVA Southeastern University, College of Dental Medicine, Fort Lauderdale, FL, USA.
| |
Collapse
|
212
|
Singh R, Pervin S, Lee SJ, Kuo A, Grijalva V, David J, Vergnes L, Reddy ST. Metabolic profiling of follistatin overexpression: a novel therapeutic strategy for metabolic diseases. Diabetes Metab Syndr Obes 2018; 11:65-84. [PMID: 29618935 PMCID: PMC5875402 DOI: 10.2147/dmso.s159315] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Follistatin (Fst) promotes brown adipocyte characteristics in adipose tissues. METHODS Abdominal fat volume (CT scan), glucose clearance (GTT test), and metabolomics analysis (mass spectrometry) of adipose tissues from Fst transgenic (Fst-Tg) and wild type (WT) control mice were analyzed. Oxygen consumption (Seahorse Analyzer) and lipidomics (gas chromatography) was analyzed in 3T3-L1 cells. RESULTS Fst-Tg mice show significant decrease in abdominal fat content, increased glucose clearance, improved plasma lipid profiles and significant changes in several conventional metabolites compared to the WT mice. Furthermore, overexpression of Fst in 3T3-L1 cells resulted in up regulation of key brown/beige markers and changes in lipidomics profiles. CONCLUSION Fst modulates key factors involved in promoting metabolic syndrome and could be used for therapeutic intervention.
Collapse
Affiliation(s)
- Rajan Singh
- Department of Obstetrics and Gynecology, UCLA School of Medicine, Los Angeles, CA, USA
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA
- Correspondence: Rajan Singh, Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, 3084 Hawkins Building, 1731 East 120 Street, Los Angeles, CA 90059, USA, Tel +1 323 563 5828, Email
| | - Shehla Pervin
- Department of Obstetrics and Gynecology, UCLA School of Medicine, Los Angeles, CA, USA
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA
| | - Se-Jin Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, CT, USA
| | - Alan Kuo
- Department of Biology, California State University Dominguez Hills, CA, USA
| | - Victor Grijalva
- Department of Molecular and Medical Pharmacology, UCLA School of Medicine, Los Angeles, CA, USA
| | - John David
- Department of Comparative Medicine, Pfizer Inc, San Diego, CA, USA
| | - Laurent Vergnes
- Department of Human Genetics, UCLA School of Medicine, Los Angeles, CA, USA
| | - Srinivasa T Reddy
- Department of Obstetrics and Gynecology, UCLA School of Medicine, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, UCLA School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
213
|
Mechanism by which arylamine N-acetyltransferase 1 ablation causes insulin resistance in mice. Proc Natl Acad Sci U S A 2017; 114:E11285-E11292. [PMID: 29237750 PMCID: PMC5748223 DOI: 10.1073/pnas.1716990115] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Insulin resistance in liver and skeletal muscle are major factors in the pathogenesis of type 2 diabetes; however, the molecular mechanism or mechanisms responsible for this phenomenon have not been established. Recently, an association of a single-nucleotide polymorphism in the human N-acetyltransferase 2 (Nat2) gene with insulin resistance in humans was found. Here, we show that the murine ortholog Nat1 knockout (KO) mice manifested whole-body insulin resistance associated with marked increases in liver and muscle lipid content. Nat1 KO mice also displayed reduced whole-body energy expenditure and reduced mitochondrial activity. Taken together, these studies demonstrate that Nat1 deletion promotes reduced mitochondrial activity and is associated with ectopic lipid-induced liver and muscle insulin resistance. A single-nucleotide polymorphism in the human arylamine N-acetyltransferase 2 (Nat2) gene has recently been identified as associated with insulin resistance in humans. To understand the cellular and molecular mechanisms by which alterations in Nat2 activity might cause insulin resistance, we examined murine ortholog Nat1 knockout (KO) mice. Nat1 KO mice manifested whole-body insulin resistance, which could be attributed to reduced muscle, liver, and adipose tissue insulin sensitivity. Hepatic and muscle insulin resistance were associated with marked increases in both liver and muscle triglyceride (TAG) and diacylglycerol (DAG) content, which was associated with increased PKCε activation in liver and increased PKCθ activation in skeletal muscle. Nat1 KO mice also displayed reduced whole-body energy expenditure and reduced mitochondrial oxygen consumption in white adipose tissue, brown adipose tissue, and hepatocytes. Taken together, these studies demonstrate that Nat1 deletion promotes reduced mitochondrial activity and is associated with ectopic lipid-induced insulin resistance. These results provide a potential genetic link among mitochondrial dysfunction with increased ectopic lipid deposition, insulin resistance, and type 2 diabetes.
Collapse
|
214
|
Xie C, Yagai T, Luo Y, Liang X, Chen T, Wang Q, Sun D, Zhao J, Ramakrishnan SK, Sun L, Jiang C, Xue X, Tian Y, Krausz KW, Patterson AD, Shah YM, Wu Y, Jiang C, Gonzalez FJ. Activation of intestinal hypoxia-inducible factor 2α during obesity contributes to hepatic steatosis. Nat Med 2017; 23:1298-1308. [PMID: 29035368 PMCID: PMC6410352 DOI: 10.1038/nm.4412] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 08/29/2017] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease is becoming the most common chronic liver disease in Western countries, and limited therapeutic options are available. Here we uncovered a role for intestinal hypoxia-inducible factor (HIF) in hepatic steatosis. Human-intestine biopsies from individuals with or without obesity revealed that intestinal HIF-2α signaling was positively correlated with body-mass index and hepatic toxicity. The causality of this correlation was verified in mice with an intestine-specific disruption of Hif2a, in which high-fat-diet-induced hepatic steatosis and obesity were substantially lower as compared to control mice. PT2385, a HIF-2α-specific inhibitor, had preventive and therapeutic effects on metabolic disorders that were dependent on intestine HIF-2α. Intestine HIF-2α inhibition markedly reduced intestine and serum ceramide levels. Mechanistically, intestine HIF-2α regulates ceramide metabolism mainly from the salvage pathway, by positively regulating the expression of Neu3, the gene encoding neuraminidase 3. These results suggest that intestinal HIF-2α could be a viable target for hepatic steatosis therapy.
Collapse
Affiliation(s)
- Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Tomoki Yagai
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Yuhong Luo
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Xianyi Liang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Tao Chen
- Department of Internal Medicine, Key Laboratory of Environment and Genes Related to Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Qiong Wang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Dongxue Sun
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jie Zhao
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sadeesh K Ramakrishnan
- Departments of Molecular & Integrative Physiology, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lulu Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Chunmei Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Xiang Xue
- Departments of Molecular & Integrative Physiology, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yuan Tian
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Yatrik M Shah
- Departments of Molecular & Integrative Physiology, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yue Wu
- Department of Internal Medicine, Key Laboratory of Environment and Genes Related to Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- These authors jointly directed this work
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
- These authors jointly directed this work
| |
Collapse
|
215
|
Macrophage VLDLR mediates obesity-induced insulin resistance with adipose tissue inflammation. Nat Commun 2017; 8:1087. [PMID: 29057873 PMCID: PMC5651811 DOI: 10.1038/s41467-017-01232-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 08/25/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity is closely associated with increased adipose tissue macrophages (ATMs), which contribute to systemic insulin resistance and altered lipid metabolism by creating a pro-inflammatory environment. Very low-density lipoprotein receptor (VLDLR) is involved in lipoprotein uptake and storage. However, whether lipid uptake via VLDLR in macrophages affects obesity-induced inflammatory responses and insulin resistance is not well understood. Here we show that elevated VLDLR expression in ATMs promotes adipose tissue inflammation and glucose intolerance in obese mice. In macrophages, VLDL treatment upregulates intracellular levels of C16:0 ceramides in a VLDLR-dependent manner, which potentiates pro-inflammatory responses and promotes M1-like macrophage polarization. Adoptive transfer of VLDLR knockout bone marrow to wild-type mice relieves adipose tissue inflammation and improves insulin resistance in diet-induced obese mice. These findings suggest that increased VLDL-VLDLR signaling in ATMs aggravates adipose tissue inflammation and insulin resistance in obesity. VLDLR regulates cellular lipoprotein uptake and storage. Here, the authors show that VLDLR, expressed on adipose tissue macrophages, is upregulated in obesity and promotes adipose tissue inflammation by upregulating ceramide production and facilitating M1-like macrophage polarization.
Collapse
|
216
|
Lee SY, Lee HY, Song JH, Kim GT, Jeon S, Song YJ, Lee JS, Hur JH, Oh HH, Park SY, Shim SM, Yoo HJ, Lee BC, Jiang XC, Choi CS, Park TS. Adipocyte-Specific Deficiency of De Novo Sphingolipid Biosynthesis Leads to Lipodystrophy and Insulin Resistance. Diabetes 2017; 66:2596-2609. [PMID: 28698261 PMCID: PMC7970771 DOI: 10.2337/db16-1232] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 06/28/2017] [Indexed: 12/16/2022]
Abstract
Sphingolipids have been implicated in the etiology of chronic metabolic diseases. Here, we investigated whether sphingolipid biosynthesis is associated with the development of adipose tissues and metabolic diseases. SPTLC2, a subunit of serine palmitoyltransferase, was transcriptionally upregulated in the adipose tissues of obese mice and in differentiating adipocytes. Adipocyte-specific SPTLC2-deficient (aSPTLC2 KO) mice had markedly reduced adipose tissue mass. Fatty acids that were destined for the adipose tissue were instead shunted to liver and caused hepatosteatosis. This impaired fat distribution caused systemic insulin resistance and hyperglycemia, indicating severe lipodystrophy. Mechanistically, sphingosine 1-phosphate (S1P) was reduced in the adipose tissues of aSPTLC2 KO mice, and this inhibited adipocyte proliferation and differentiation via the downregulation of S1P receptor 1 and decreased activity of the peroxisome proliferator-activator receptor γ. In addition, downregulation of SREBP (sterol regulatory element-binding protein)-1c prevented adipogenesis of aSPTLC2 KO adipocytes. Collectively, our observations suggest that the tight regulation of de novo sphingolipid biosynthesis and S1P signaling plays an important role in adipogenesis and hepatosteatosis.
Collapse
Affiliation(s)
- Su-Yeon Lee
- Department of Life Science, Gachon University, Sungnam, Korea
| | - Hui-Young Lee
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, Korea
| | - Jae-Hwi Song
- Department of Life Science, Gachon University, Sungnam, Korea
| | - Goon-Tae Kim
- Department of Life Science, Gachon University, Sungnam, Korea
| | - Suwon Jeon
- Department of Life Science, Gachon University, Sungnam, Korea
| | - Yoo-Jeong Song
- Department of Life Science, Gachon University, Sungnam, Korea
| | - Jae Sung Lee
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, Korea
| | - Jang-Ho Hur
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, Korea
| | - Hyun Hee Oh
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, Korea
| | - Shi-Young Park
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, Korea
| | - Soon-Mi Shim
- Department of Food Science and Technology, Sejong University, Seoul, Korea
| | - Hyun Joo Yoo
- Biomedical Research Center, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Byung Cheon Lee
- College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Xian-Cheng Jiang
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Cheol Soo Choi
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, Korea
- Endocrinology, Internal Medicine, Gachon University Gil Medical Center, Incheon, Korea
| | - Tae-Sik Park
- Department of Life Science, Gachon University, Sungnam, Korea
| |
Collapse
|
217
|
Adipose Tissue Function and Expandability as Determinants of Lipotoxicity and the Metabolic Syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:161-196. [PMID: 28585199 DOI: 10.1007/978-3-319-48382-5_7] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The adipose tissue organ is organised as distinct anatomical depots located all along the body axis and it is constituted of three different types of adipocytes : white, beige and brown which are integrated with vascular, immune, neural and extracellular stroma cells. These distinct adipocytes serve different specialised functions. The main function of white adipocytes is to ensure healthy storage of excess nutrients/energy and its rapid mobilisation to supply the demand of energy imposed by physiological cues in other organs, whereas brown and beige adipocytes are designed for heat production through uncoupling lipid oxidation from energy production. The concert action of the three type of adipocytes/tissues has been reported to ensure an optimal metabolic status in rodents. However, when one or multiple of these adipose depots become dysfunctional as a consequence of sustained lipid/nutrient overload, then insulin resistance and associated metabolic complications ensue. These metabolic alterations negatively affects the adipose tissue functionality and compromises global metabolic homeostasis. Optimising white adipose tissue expandability and its functional metabolic flexibility and/or promoting brown/beige mediated thermogenic activity counteracts obesity and its associated lipotoxic metabolic effects. The development of these therapeutic approaches requires a deep understanding of adipose tissue in all broad aspects. In this chapter we will discuss the characteristics of the different adipose tissue depots with respect to origins and precursors recruitment, plasticity, cellular composition and expandability capacity as well as molecular and metabolic signatures in both physiological and pathophysiological conditions.
Collapse
|
218
|
Davis AN, Clegg JL, Perry CA, McFadden JW. Nutrient Restriction Increases Circulating and Hepatic Ceramide in Dairy Cows Displaying Impaired Insulin Tolerance. Lipids 2017; 52:771-780. [PMID: 28836149 DOI: 10.1007/s11745-017-4287-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 08/10/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Amanda N. Davis
- ; Division of Animal and Nutritional Sciences; West Virginia University; Morgantown 26505 USA
| | - J. L. Clegg
- ; Division of Animal and Nutritional Sciences; West Virginia University; Morgantown 26505 USA
| | - C. A. Perry
- ; Division of Animal and Nutritional Sciences; West Virginia University; Morgantown 26505 USA
| | - J. W. McFadden
- ; Division of Animal and Nutritional Sciences; West Virginia University; Morgantown 26505 USA
- ; Cornell University; 264 Morrison Hall Ithaca NY 14853 USA
| |
Collapse
|
219
|
Liao W, Liu H, Zhang Y, Jung JH, Chen J, Su X, Kim YC, Flores ER, Wang SM, Czarny-Ratajczak M, Li W, Zeng SX, Lu H. Ccdc3: A New P63 Target Involved in Regulation Of Liver Lipid Metabolism. Sci Rep 2017; 7:9020. [PMID: 28827783 PMCID: PMC5566403 DOI: 10.1038/s41598-017-09228-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 07/17/2017] [Indexed: 12/18/2022] Open
Abstract
TAp63, a member of the p53 family, has been shown to regulate energy metabolism. Here, we report coiled coil domain-containing 3 (CCDC3) as a new TAp63 target. TAp63, but not ΔNp63, p53 or p73, upregulates CCDC3 expression by directly binding to its enhancer region. The CCDC3 expression is markedly reduced in TAp63-null mouse embryonic fibroblasts and brown adipose tissues and by tumor necrosis factor alpha that reduces p63 transcriptional activity, but induced by metformin, an anti-diabetic drug that activates p63. Also, the expression of CCDC3 is positively correlated with TAp63 levels, but conversely with ΔNp63 levels, during adipocyte differentiation. Interestingly, CCDC3, as a secreted protein, targets liver cancer cells and increases long chain polyunsaturated fatty acids, but decreases ceramide in the cells. CCDC3 alleviates glucose intolerance, insulin resistance and steatosis formation in transgenic CCDC3 mice on high-fat diet (HFD) by reducing the expression of hepatic PPARγ and its target gene CIDEA as well as other genes involved in de novo lipogenesis. Similar results are reproduced by hepatic expression of ectopic CCDC3 in mice on HFD. Altogether, these results demonstrate that CCDC3 modulates liver lipid metabolism by inhibiting liver de novo lipogenesis as a downstream player of the p63 network.
Collapse
Affiliation(s)
- Wenjuan Liao
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Hongbing Liu
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Yiwei Zhang
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Ji Hoon Jung
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Jiaxiang Chen
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,Department of Physiology, Jiangxi Medical College of Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Xiaohua Su
- Department of Molecular Oncology, Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Yeong C Kim
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, 68198, NE, USA
| | - Elsa R Flores
- Department of Molecular Oncology, Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - San Ming Wang
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, 68198, NE, USA.,Faculty of Health Sciences, University of Macau, Macau, China
| | - Malwina Czarny-Ratajczak
- Department of Medicine, Center for Aging, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Wen Li
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yatsen University, Guangzhou, Guangdong, 510080, P.R. China
| | - Shelya X Zeng
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| | - Hua Lu
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
220
|
Untargeted metabolomics identifies a plasma sphingolipid-related signature associated with lifestyle intervention in prepubertal children with obesity. Int J Obes (Lond) 2017; 42:72-78. [PMID: 28947825 DOI: 10.1038/ijo.2017.201] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/25/2017] [Accepted: 08/02/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Childhood obesity is a strong risk factor for adult obesity and metabolic diseases, including type 2 diabetes and cardiovascular disease. Early lifestyle intervention in children with obesity reduces future disease risk. The objective of this study is to identify metabolic signatures associated with lifestyle intervention in prepubertal children with obesity. METHODS Thirty-five prepubertal children (7-10 years) with obesity (body mass index (BMI)>2 standard deviations) were enrolled in the study and participated in a 6-month-long lifestyle intervention program. Physiological and biochemical data and blood samples were collected both at baseline and after the intervention. A liquid chromatography-mass spectrometry (LC-MS)-based metabolomics approach was applied to obtain a comprehensive profiling of plasma samples, identifying 2581 distinct metabolite. Principal component analysis (PCA) was performed to consolidate all features into 8 principal components. Associations between metabolites and physiological and biochemical variables were investigated. RESULTS The intervention program significantly decreased mean (95% CI) BMI standard deviation score from 3.56 (3.29-3.84) to 3.11 (2.88-3.34) (P<0.001). PCA identified one component (PC1) significantly altered by the intervention (Bonferroni adjusted P=0.008). A sphingolipid metabolism-related signature was identified as the major contributor to PC1. Sphingolipid metabolites were decreased by the intervention, and included multiple sphingomyelin, ceramide, glycosylsphingosine and sulfatide species. Changes in several sphingolipid metabolites were associated with intervention-induced improvements in HbA1c levels. CONCLUSIONS Decreased circulating sphingolipid-related metabolites were associated with lifestyle intervention in prepubertal children with obesity, and correlated to improvements in HbA1c.
Collapse
|
221
|
Chen TC, Benjamin DI, Kuo T, Lee RA, Li ML, Mar DJ, Costello DE, Nomura DK, Wang JC. The glucocorticoid-Angptl4-ceramide axis induces insulin resistance through PP2A and PKCζ. Sci Signal 2017; 10:eaai7905. [PMID: 28743803 PMCID: PMC6218395 DOI: 10.1126/scisignal.aai7905] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chronic glucocorticoid exposure is associated with the development of insulin resistance. We showed that glucocorticoid-induced insulin resistance was attenuated upon ablation of Angptl4, a glucocorticoid target gene encoding the secreted protein angiopoietin-like 4, which mediates glucocorticoid-induced lipolysis in white adipose tissue. Through metabolomic profiling, we revealed that glucocorticoid treatment increased hepatic ceramide concentrations by inducing enzymes in the ceramide synthetic pathway in an Angptl4-dependent manner. Angptl4 was also required for glucocorticoids to stimulate the activities of the downstream effectors of ceramide, protein phosphatase 2A (PP2A) and protein kinase Cζ (PKCζ). We further showed that knockdown of PP2A or inhibition of PKCζ or ceramide synthesis prevented glucocorticoid-induced glucose intolerance in wild-type mice. Moreover, the inhibition of PKCζ or ceramide synthesis did not further improve glucose tolerance in Angptl4-/- mice, suggesting that these molecules were major downstream effectors of Angptl4. Overall, our study demonstrates the key role of Angptl4 in glucocorticoid-augmented hepatic ceramide production that induces whole-body insulin resistance.
Collapse
Affiliation(s)
- Tzu-Chieh Chen
- Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720-3104, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720-3104, USA
| | - Daniel I Benjamin
- Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720-3104, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720-3104, USA
| | - Taiyi Kuo
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720-3104, USA
- Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720-3104, USA
| | - Rebecca A Lee
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720-3104, USA
- Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720-3104, USA
| | - Mei-Lan Li
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720-3104, USA
| | - Darryl J Mar
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720-3104, USA
| | - Damian E Costello
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720-3104, USA
- Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720-3104, USA
| | - Daniel K Nomura
- Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720-3104, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720-3104, USA
- Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720-3104, USA
- Departments of Chemistry and Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720-3104, USA
| | - Jen-Chywan Wang
- Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720-3104, USA.
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720-3104, USA
- Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720-3104, USA
| |
Collapse
|
222
|
The expanding role of sphingolipids in lipid droplet biogenesis. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1155-1165. [PMID: 28743537 DOI: 10.1016/j.bbalip.2017.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 01/17/2023]
Abstract
Sphingolipids are a diverse class of lipids that have regulatory, structural, and metabolic functions. Although chemically distinct from the neutral lipids and the glycerophospholipids, which are the main lipid components of the lipid droplets, sphingolipids have nonetheless been shown to influence lipid droplet formation. The goal of this article is to review the available information and provide a cohesive picture of the role sphingolipids play in lipid droplet biogenesis. The following topics are discussed: (i) the abundance of sphingolipids in lipid droplets and their functional significance; (ii) cross-talk between the synthetic pathways of sphingolipids, glycerophospholipids, and neutral lipids; (iii) the impact of bioactive sphingolipids on TAG synthesis and degradation; (iv) interactions between sphingolipids and other lipid droplet components, like cholesterol esters and proteins; (v) inhibition/genetic deletion of specific sphingolipid metabolic enzymes and the resulting effects on lipid droplet formation in mouse models. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
|
223
|
Sharma AX, Holland WL. Adiponectin and its Hydrolase-Activated Receptors. JOURNAL OF NATURE AND SCIENCE 2017; 3:e396. [PMID: 28758149 PMCID: PMC5531184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The relevance of adiponectin to insulin sensitivity has been elucidated over the last two decades. As a promoter of ceramide degradation, it works through its cognate receptors, AdipoR1 and AdipoR2, to alter bioactive sphingolipid species. Adiponectin diminishes the accumulation of ceramide, a lipid metabolite which can play a causal role in obesity-induced insulin resistance. Concurrently, adiponectin stimulates the production of sphingosine-1-phosphate (S1P), a cyto-protective molecule that accentuates adiponectin's positive metabolic effects. This review focuses on recent work that solidifies knowledge of the adiponectin signaling pathway, gives new insight into some notable characteristics of adiponectin's receptors, and most importantly, affirms adiponectin receptor agonism as a viable therapeutic tool to combat elevated ceramide levels and improve insulin sensitivity in obese patients with type II diabetes.
Collapse
Affiliation(s)
| | - William L. Holland
- Corresponding Author: William L. Holland. Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390-8549, USA. Tel: 214-648-2566; Fax: 214-648-8720.
| |
Collapse
|
224
|
Petersen MC, Shulman GI. Roles of Diacylglycerols and Ceramides in Hepatic Insulin Resistance. Trends Pharmacol Sci 2017; 38:649-665. [PMID: 28551355 DOI: 10.1016/j.tips.2017.04.004] [Citation(s) in RCA: 245] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 12/22/2022]
Abstract
Although ample evidence links hepatic lipid accumulation with hepatic insulin resistance, the mechanistic basis of this association is incompletely understood and controversial. Diacylglycerols (DAGs) and ceramides have emerged as the two best-studied putative mediators of lipid-induced hepatic insulin resistance. Both lipids were first associated with insulin resistance in skeletal muscle and were subsequently hypothesized to mediate insulin resistance in the liver. However, the putative roles for DAGs and ceramides in hepatic insulin resistance have proved more complex than originally imagined, with various genetic and pharmacologic manipulations yielding a vast and occasionally contradictory trove of data to sort. In this review we examine the state of this field, turning a critical eye toward both DAGs and ceramides as putative mediators of lipid-induced hepatic insulin resistance.
Collapse
Affiliation(s)
- Max C Petersen
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gerald I Shulman
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
225
|
Taylor OJ, Thatcher MO, Carr ST, Gibbs JL, Trumbull AM, Harrison ME, Winden DR, Pearson MJ, Tippetts TS, Holland WL, Reynolds PR, Bikman BT. High-Mobility Group Box 1 Disrupts Metabolic Function with Cigarette Smoke Exposure in a Ceramide-Dependent Manner. Int J Mol Sci 2017; 18:E1099. [PMID: 28531105 PMCID: PMC5455007 DOI: 10.3390/ijms18051099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 05/16/2017] [Accepted: 05/18/2017] [Indexed: 11/23/2022] Open
Abstract
We have previously found that cigarette smoke disrupts metabolic function, in part, by increasing muscle ceramide accrual. To further our understanding of this, we sought to determine the role of the cytokine high-mobility group box 1 (HMGB1), which is increased with smoke exposure, in smoke-induced muscle metabolic perturbations. To test this theory, we determined HMGB1 from lungs of human smokers, as well as from lung cells from mice exposed to cigarette smoke. We also treated cells and mice directly with HMGB1, in the presence or absence of myriocin, an inhibitor of serine palmitoyltransferase, the rate-limiting enzyme in ceramide biosynthesis. Outcomes included assessments of insulin resistance and muscle mitochondrial function. HMGB1 was significantly increased in both human lungs and rodent alveolar macrophages. Further testing revealed that HMGB1 treatment elicited a widespread increase in ceramide species and reduction in myotube mitochondrial respiration, an increase in reactive oxygen species, and reduced insulin-stimulated Akt phosphorylation. Inhibition of ceramide biosynthesis with myriocin was protective. In mice, by comparing treatments of HMGB1 injections with or without myriocin, we found that HMGB1 injections resulted in increased muscle ceramides, especially C16 and C24, which were necessary for reduced muscle mitochondrial respiration and compromised insulin and glucose tolerance. In conclusion, HMGB1 may be a necessary intermediate in the ceramide-dependent metabolic consequences of cigarette smoke exposure.
Collapse
Affiliation(s)
- Oliver J Taylor
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA.
| | - Mikayla O Thatcher
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA.
| | - Sheryl T Carr
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA.
| | - Jonathan L Gibbs
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA.
| | - Annie M Trumbull
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA.
| | - Mitchell E Harrison
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA.
| | - Duane R Winden
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT 84095, USA.
| | - Mackenzie J Pearson
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390; USA.
| | - Trevor S Tippetts
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390; USA.
| | - William L Holland
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390; USA.
| | - Paul R Reynolds
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA.
| | - Benjamin T Bikman
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
226
|
Rico JE, Saed Samii S, Mathews AT, Lovett J, Haughey NJ, McFadden JW. Temporal changes in sphingolipids and systemic insulin sensitivity during the transition from gestation to lactation. PLoS One 2017; 12:e0176787. [PMID: 28486481 PMCID: PMC5423608 DOI: 10.1371/journal.pone.0176787] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 04/17/2017] [Indexed: 12/17/2022] Open
Abstract
Reduced insulin action develops naturally during the peripartum to ensure maternal nutrient delivery to the fetus and neonate. However, increased insulin resistance can facilitate excessive lipolysis which in turn promotes metabolic disease in overweight dairy cattle. Increased fatty acid availability favors the accumulation of the sphingolipid ceramide and is implicated in the pathogenesis of insulin resistance, however, the relationship between sphingolipid metabolism and insulin resistance during the peripartum remains largely unknown. Our objectives were to characterize temporal responses in plasma and tissue sphingolipids in lean and overweight peripartal cows and to establish the relationships between sphingolipid supply and lipolysis, hepatic lipid deposition, and systemic insulin action. Twenty-one multiparous lean and overweight Holstein cows were enrolled in a longitudinal study spanning the transition from gestation to lactation (d -21 to 21, relative to parturition). Plasma, liver, and skeletal muscle samples were obtained, and sphingolipids were profiled using LC/MS/MS. Insulin sensitivity was assessed utilizing intravenous insulin and glucose challenges. Our results demonstrated the following: first, insulin resistance develops postpartum concurrently with increased lipolysis and hepatic lipid accumulation; second, ceramides and glycosylated ceramides accumulate during the transition from gestation to lactation and are further elevated in overweight cows; third, ceramide accrual is associated with lipolysis and liver lipid accumulation, and C16:0- and C24:0-ceramide are inversely associated with systemic insulin sensitivity postpartum; fourth, plasma sphingomyelin, a potential source of ceramides reaches a nadir at parturition and is closely associated with feed intake; fifth, select sphingomyelins are lower in the plasma of overweight cows during the peripartal period. Our results demonstrate that dynamic changes occur in peripartal sphingolipids that are influenced by adiposity, and are associated with the onset of peripartal insulin resistance. These observations are in agreement with a putative potential role for sphingolipids in facilitating the physiological adaptations of peripartum.
Collapse
Affiliation(s)
- J. Eduardo Rico
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, West Virginia, United States of America
| | - Sina Saed Samii
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, West Virginia, United States of America
| | - Alice T. Mathews
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, West Virginia, United States of America
| | - Jacqueline Lovett
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Norman J. Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Joseph W. McFadden
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, West Virginia, United States of America
| |
Collapse
|
227
|
Mauer AS, Hirsova P, Maiers JL, Shah VH, Malhi H. Inhibition of sphingosine 1-phosphate signaling ameliorates murine nonalcoholic steatohepatitis. Am J Physiol Gastrointest Liver Physiol 2017; 312:G300-G313. [PMID: 28039158 PMCID: PMC5401989 DOI: 10.1152/ajpgi.00222.2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 11/28/2016] [Accepted: 12/11/2016] [Indexed: 01/31/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a lipotoxic disorder, wherein proinflammatory lipids, such as ceramide and its derivative sphingosine 1-phosphate (S1P), contribute to macrophage-associated liver inflammation. For example, we have previously demonstrated a role for S1P in steatotic hepatocyte-derived S1P-enriched extracellular vesicles in macrophage chemotaxis in vitro. Therefore, we hypothesized that FTY720, an S1P antagonist, would ameliorate NASH by inhibiting proinflammatory monocyte chemotaxis. To test our hypothesis, NASH was established in C57BL/6 male mice by feeding a diet high in fructose, saturated fat, and cholesterol for 22 wk. Then mice received daily intraperitoneal injections of FTY720 for 2 wk before analysis of liver injury, inflammation, and fibrosis. FTY720-treated mice with NASH demonstrated improved liver histology with a significant reduction in hepatocyte ballooning and inflammatory foci. Hepatomegaly was reversed, and liver triglycerides were reduced following FTY720 administration to mice with NASH. Correspondingly, serum ALT levels, hepatic inflammatory macrophage accumulation, and the expression of Ly6C in recruited myeloid cells was reduced in FTY720-treated mice. Hepatic collagen accumulation and expression of α-smooth muscle actin were significantly lowered as well. Body composition, energy consumption and utilization, and hepatic sphingolipid composition remained unchanged following FTY720 administration. FTY720 ameliorates murine nonalcoholic steatohepatitis. Reduction in liver injury and inflammation is associated with a reduction in hepatic macrophage accumulation, likely due to dampened recruitment of circulating myeloid cells into the liver. Nonalcoholic steatohepatitis may be a novel indication for the therapeutic use of FTY720.NEW & NOTEWORTHY There are no approved pharmacologic therapies for nonalcoholic steatohepatitis (NASH), the leading cause of chronic liver disease worldwide. This study describes the use of FTY720, a novel small molecule, for the amelioration of NASH in a mouse model. We demonstrate that 2-wk administration of FTY720 to mice with NASH led to a reduction in liver injury, inflammation, and fibrosis. These data provide a preclinical rationale for studying this drug in human NASH.
Collapse
Affiliation(s)
- Amy S Mauer
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Petra Hirsova
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Jessica L Maiers
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
228
|
Xie C, Jiang C, Shi J, Gao X, Sun D, Sun L, Wang T, Takahashi S, Anitha M, Krausz KW, Patterson AD, Gonzalez FJ. An Intestinal Farnesoid X Receptor-Ceramide Signaling Axis Modulates Hepatic Gluconeogenesis in Mice. Diabetes 2017; 66:613-626. [PMID: 28223344 PMCID: PMC5319721 DOI: 10.2337/db16-0663] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 10/29/2016] [Indexed: 12/12/2022]
Abstract
Increasing evidence supports the view that intestinal farnesoid X receptor (FXR) is involved in glucose tolerance and that FXR signaling can be profoundly impacted by the gut microbiota. Selective manipulation of the gut microbiota-FXR signaling axis was reported to significantly impact glucose intolerance, but the precise molecular mechanism remains largely unknown. Here, caffeic acid phenethyl ester (CAPE), an over-the-counter dietary supplement and an inhibitor of bacterial bile salt hydrolase, increased levels of intestinal tauro-β-muricholic acid, which selectively suppresses intestinal FXR signaling. Intestinal FXR inhibition decreased ceramide levels by suppressing expression of genes involved in ceramide synthesis specifically in the intestinal ileum epithelial cells. The lower serum ceramides mediated decreased hepatic mitochondrial acetyl-CoA levels and pyruvate carboxylase (PC) activities and attenuated hepatic gluconeogenesis, independent of body weight change and hepatic insulin signaling in vivo; this was reversed by treatment of mice with ceramides or the FXR agonist GW4064. Ceramides substantially attenuated mitochondrial citrate synthase activities primarily through the induction of endoplasmic reticulum stress, which triggers increased hepatic mitochondrial acetyl-CoA levels and PC activities. These results reveal a mechanism by which the dietary supplement CAPE and intestinal FXR regulates hepatic gluconeogenesis and suggest that inhibiting intestinal FXR is a strategy for treating hyperglycemia.
Collapse
Affiliation(s)
- Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Jingmin Shi
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Xiaoxia Gao
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Dongxue Sun
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lulu Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Ting Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Shogo Takahashi
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Mallappa Anitha
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Andrew D Patterson
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
229
|
Reibe-Pal S, Febbraio MA. Adiponectin serenades ceramidase to improve metabolism. Mol Metab 2017; 6:233-235. [PMID: 28271029 PMCID: PMC5324015 DOI: 10.1016/j.molmet.2017.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 01/25/2017] [Indexed: 01/14/2023] Open
Affiliation(s)
- Saskia Reibe-Pal
- Cellular & Molecular Metabolism Laboratory, Diabetes & Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia
| | - Mark A Febbraio
- Cellular & Molecular Metabolism Laboratory, Diabetes & Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia.
| |
Collapse
|
230
|
Holland WL, Xia JY, Johnson JA, Sun K, Pearson MJ, Sharma AX, Quittner-Strom E, Tippetts TS, Gordillo R, Scherer PE. Inducible overexpression of adiponectin receptors highlight the roles of adiponectin-induced ceramidase signaling in lipid and glucose homeostasis. Mol Metab 2017; 6:267-275. [PMID: 28271033 PMCID: PMC5323887 DOI: 10.1016/j.molmet.2017.01.002] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 12/27/2016] [Accepted: 01/05/2017] [Indexed: 02/06/2023] Open
Abstract
Objective Adiponectin and the signaling induced by its cognate receptors, AdipoR1 and AdipoR2, have garnered attention for their ability to promote insulin sensitivity and oppose steatosis. Activation of these receptors promotes the deacylation of ceramide, a lipid metabolite that appears to play a causal role in impairing insulin signaling. Methods Here, we have developed transgenic mice that overexpress AdipoR1 or AdipoR2 under the inducible control of a tetracycline response element. These represent the first inducible genetic models that acutely manipulate adiponectin receptor signaling in adult mouse tissues, which allows us to directly assess AdipoR signaling on glucose and lipid metabolism. Results Overexpression of either adiponectin receptor isoform in the adipocyte or hepatocyte is sufficient to enhance ceramidase activity, whole body glucose metabolism, and hepatic insulin sensitivity, while opposing hepatic steatosis. Importantly, metabolic improvements fail to occur in an adiponectin knockout background. When challenged with a leptin-deficient genetic model of type 2 diabetes, AdipoR2 expression in adipose or liver is sufficient to reverse hyperglycemia and glucose intolerance. Conclusion These observations reveal that adiponectin is critical for AdipoR-induced ceramidase activation which enhances hepatic glucose and lipid metabolism via rapidly acting “cross-talk” between liver and adipose tissue sphingolipids. Adiponectin receptor signaling in adipose prompts beneficial effects on whole-body glucose and lipid metabolism. The small molecule adiponectin receptor antagonist AdipoRon lowers hepatic ceramides. Depletion of ceramides in adipocytes results in diminished hepatic ceramide accumulation. Depletion of ceramides in hepatocytes results in diminished adipose sphingolipid accumulation. Adiponectin is essential for the beneficial effects of adiponectin receptors on glucose, ceramide, and lipid metabolism.
Collapse
Affiliation(s)
- William L Holland
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA.
| | - Jonathan Y Xia
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Joshua A Johnson
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Kai Sun
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Mackenzie J Pearson
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Ankit X Sharma
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Ezekiel Quittner-Strom
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Trevor S Tippetts
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA; Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| |
Collapse
|
231
|
Chaurasia B, Kaddai VA, Lancaster GI, Henstridge DC, Sriram S, Galam DLA, Gopalan V, Prakash KNB, Velan SS, Bulchand S, Tsong TJ, Wang M, Siddique MM, Yuguang G, Sigmundsson K, Mellet NA, Weir JM, Meikle PJ, Bin M Yassin MS, Shabbir A, Shayman JA, Hirabayashi Y, Shiow SATE, Sugii S, Summers SA. Adipocyte Ceramides Regulate Subcutaneous Adipose Browning, Inflammation, and Metabolism. Cell Metab 2016; 24:820-834. [PMID: 27818258 DOI: 10.1016/j.cmet.2016.10.002] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 06/15/2016] [Accepted: 10/05/2016] [Indexed: 01/23/2023]
Abstract
Adipocytes package incoming fatty acids into triglycerides and other glycerolipids, with only a fraction spilling into a parallel biosynthetic pathway that produces sphingolipids. Herein, we demonstrate that subcutaneous adipose tissue of type 2 diabetics contains considerably more sphingolipids than non-diabetic, BMI-matched counterparts. Whole-body and adipose tissue-specific inhibition/deletion of serine palmitoyltransferase (Sptlc), the first enzyme in the sphingolipid biosynthesis cascade, in mice markedly altered adipose morphology and metabolism, particularly in subcutaneous adipose tissue. The reduction in adipose sphingolipids increased brown and beige/brite adipocyte numbers, mitochondrial activity, and insulin sensitivity. The manipulation also increased numbers of anti-inflammatory M2 macrophages in the adipose bed and induced secretion of insulin-sensitizing adipokines. By comparison, deletion of serine palmitoyltransferase from macrophages had no discernible effects on metabolic homeostasis or adipose function. These data indicate that newly synthesized adipocyte sphingolipids are nutrient signals that drive changes in the adipose phenotype to influence whole-body energy expenditure and nutrient metabolism.
Collapse
Affiliation(s)
- Bhagirath Chaurasia
- Laboratory of Translational Metabolic Health, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia.
| | - Vincent Andre Kaddai
- Laboratory of Translational Metabolic Health, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia
| | - Graeme Iain Lancaster
- Laboratory of Translational Metabolic Health, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia
| | - Darren C Henstridge
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia
| | - Sandhya Sriram
- Fat Metabolism and Stem Cell Group, Singapore Bioimaging Consortium, Singapore 138667, Singapore
| | - Dwight Lark Anolin Galam
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Graduate Medical School, Singapore 169547, Singapore
| | - Venkatesh Gopalan
- Laboratory of Molecular Imaging, Singapore Bioimaging Consortium, Singapore 138667, Singapore
| | - K N Bhanu Prakash
- Laboratory of Molecular Imaging, Singapore Bioimaging Consortium, Singapore 138667, Singapore
| | - S Sendhil Velan
- Laboratory of Molecular Imaging, Singapore Bioimaging Consortium, Singapore 138667, Singapore
| | - Sarada Bulchand
- Tata Institute of Fundamental Research, Navy Nagar, Mumbai 400005, India
| | - Teh Jing Tsong
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Mei Wang
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | | | - Guan Yuguang
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Graduate Medical School, Singapore 169547, Singapore
| | - Kristmundur Sigmundsson
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Graduate Medical School, Singapore 169547, Singapore
| | - Natalie A Mellet
- Metabolomics Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia
| | - Jacquelyn M Weir
- Metabolomics Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia
| | - M Shabeer Bin M Yassin
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Asim Shabbir
- Department of Surgery, National University of Singapore, Singapore 117599, Singapore
| | - James A Shayman
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | - Sue-Anne Toh Ee Shiow
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Shigeki Sugii
- Fat Metabolism and Stem Cell Group, Singapore Bioimaging Consortium, Singapore 138667, Singapore; Program in Cardiovascular and Metabolic Disorders, Duke-NUS Graduate Medical School, Singapore 169547, Singapore
| | - Scott A Summers
- Laboratory of Translational Metabolic Health, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia
| |
Collapse
|
232
|
Gonzalez FJ, Jiang C, Patterson AD. An Intestinal Microbiota-Farnesoid X Receptor Axis Modulates Metabolic Disease. Gastroenterology 2016; 151:845-859. [PMID: 27639801 PMCID: PMC5159222 DOI: 10.1053/j.gastro.2016.08.057] [Citation(s) in RCA: 255] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/02/2016] [Accepted: 08/12/2016] [Indexed: 02/07/2023]
Abstract
The gut microbiota is associated with metabolic diseases including obesity, insulin resistance, and nonalcoholic fatty liver disease, as shown by correlative studies and by transplant of microbiota from obese humans and mice into germ-free mice. Modification of the microbiota by treatment of high-fat diet (HFD)-fed mice with tempol or antibiotics resulted in decreased adverse metabolic phenotypes. This was owing to lower levels of the genera Lactobacillus and decreased bile salt hydrolase (BSH) activity. The decreased BSH resulted in increased levels of tauro-β-muricholic acid (MCA), a substrate of BSH and a potent farnesoid X receptor (FXR) antagonist. Mice lacking expression of FXR in the intestine were resistant to HFD-induced obesity, insulin resistance, and nonalcoholic fatty liver disease, thus confirming that intestinal FXR is involved in the potentiation of metabolic disease. A potent intestinal FXR antagonist, glycine-β-MCA (Gly-MCA), which is resistant to BSH, was developed, which, when administered to HFD-treated mice, mimics the effect of the altered microbiota on HFD-induced metabolic disease. Gly-MCA had similar effects on genetically obese leptin-deficient mice. The decrease in adverse metabolic phenotype by tempol, antibiotics, and Gly-MCA was caused by decreased serum ceramides. Mice lacking FXR in the intestine also have lower serum ceramide levels, and are resistant to HFD-induced metabolic disease, and this was reversed by injection of C16:0 ceramide. In mouse ileum, because of the presence of endogenous FXR agonists produced in the liver, FXR target genes involved in ceramide synthesis are activated and when Gly-MCA is administered they are repressed, which likely accounts for the decrease in serum ceramides. These studies show that ceramides produced in the ileum under control of FXR influence metabolic diseases.
Collapse
Affiliation(s)
- Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, P. R. China
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802
| |
Collapse
|
233
|
Camell C, Goldberg E, Dixit VD. Regulation of Nlrp3 inflammasome by dietary metabolites. Semin Immunol 2016; 27:334-42. [PMID: 26776831 DOI: 10.1016/j.smim.2015.10.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 10/14/2015] [Accepted: 10/19/2015] [Indexed: 12/21/2022]
Abstract
The bidirectional communication between innate immune cells and energy metabolism is now widely appreciated to regulate homeostasis as well as chronic diseases that emerge from dysregulated inflammation. Macronutrients-derived from diet or endogenous pathways that generate and divert metabolites into energetic or biosynthetic pathways – regulate the initiation, duration and cessation of the inflammatory response. The NLRP3 inflammasome is an important innate sensor of structurally diverse metabolic damage-associated molecular patterns (DAMPs) that has been implicated in a wide range of inflammatory disorders associated with caloric excess, adiposity and aging. Understanding the regulators of immune-metabolic interactions and their contribution towards chronic disease mechanisms, therefore, has the potential to reduce disease pathology, improve quality of life in elderly and promote the extension of healthspan. Just as specialized subsets of immune cells dampen inflammation through the production of negative regulatory cytokines; specific immunoregulatory metabolites can deactivate inflammasome-mediated immune activation. Here, we highlight the role of energy substrates, alternative fuels and metabolic DAMPs in the regulation of the NLRP3 inflammasome and discuss potential dietary interventions that may impact sterile inflammatory disease.
Collapse
|
234
|
Zlobine I, Gopal K, Ussher JR. Lipotoxicity in obesity and diabetes-related cardiac dysfunction. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1555-68. [DOI: 10.1016/j.bbalip.2016.02.011] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 02/15/2016] [Indexed: 12/11/2022]
|
235
|
Hirsova P, Ibrabim SH, Gores GJ, Malhi H. Lipotoxic lethal and sublethal stress signaling in hepatocytes: relevance to NASH pathogenesis. J Lipid Res 2016; 57:1758-1770. [PMID: 27049024 PMCID: PMC5036373 DOI: 10.1194/jlr.r066357] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/05/2016] [Indexed: 12/12/2022] Open
Abstract
The accumulation of lipids is a histologic and biochemical hallmark of obesity-associated nonalcoholic fatty liver disease (NAFLD). A subset of NALFD patients develops progressive liver disease, termed nonalcoholic steatohepatitis, which is characterized by hepatocellular apoptosis and innate immune system-mediated inflammation. These responses are orchestrated by signaling pathways that can be activated by lipids, directly or indirectly. In this review, we discuss palmitate- and lysophosphatidylcholine (LPC)-induced upregulation of p53-upregulated modulator of apoptosis and cell-surface expression of the death receptor TNF-related apoptosis-inducing ligand receptor 2. Next, we review the activation of stress-induced kinases, mixed lineage kinase 3, and c-Jun N-terminal kinase, and the activation of endoplasmic reticulum stress response and its downstream proapoptotic effector, CAAT/enhancer binding homologous protein, by palmitate and LPC. Moreover, the activation of these stress signaling pathways is linked to the release of proinflammatory, proangiogenic, and profibrotic extracellular vesicles by stressed hepatocytes. This review discusses the signaling pathways induced by lethal and sublethal lipid overload that contribute to the pathogenesis of NAFLD.
Collapse
Affiliation(s)
- Petra Hirsova
- Divisions of Gastroenterology and Hepatology Mayo Clinic, Rochester, MN 55905
| | - Samar H Ibrabim
- Pediatric Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Gregory J Gores
- Divisions of Gastroenterology and Hepatology Mayo Clinic, Rochester, MN 55905.
| | - Harmeet Malhi
- Divisions of Gastroenterology and Hepatology Mayo Clinic, Rochester, MN 55905.
| |
Collapse
|
236
|
Fabbri E, Yang A, Simonsick EM, Chia CW, Zoli M, Haughey NJ, Mielke MM, Ferrucci L, Coen PM. Circulating ceramides are inversely associated with cardiorespiratory fitness in participants aged 54-96 years from the Baltimore Longitudinal Study of Aging. Aging Cell 2016; 15:825-31. [PMID: 27135629 PMCID: PMC5013023 DOI: 10.1111/acel.12491] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2016] [Indexed: 01/25/2023] Open
Abstract
Cardiorespiratory fitness (VO2 peak) declines with age and is an independent risk factor for morbidity and mortality in older adults. Identifying biomarkers of low fitness may provide insight for why some individuals experience an accelerated decline of aerobic capacity and may serve as clinically valuable prognostic indicators of cardiovascular health. We investigated the relationship between circulating ceramides and VO2 peak in 443 men and women (mean age of 69) enrolled in the Baltimore Longitudinal Study of Aging (BLSA). Individual species of ceramide were quantified by HPLC–tandem mass spectrometry. VO2 peak was measured by a graded treadmill test. We applied multiple regression models to test the associations between ceramide species and VO2 peak, while adjusting for age, sex, blood pressure, serum LDL, HDL, triglycerides, and other covariates. We found that higher levels of circulating C18:0, C20:0, C24:1 ceramides and C20:0 dihydroceramides were strongly associated with lower aerobic capacity (P < 0.001, P < 0.001, P = 0.018, and P < 0.001, respectively). The associations held true for both sexes (with men having a stronger association than women, P value for sex interaction <0.05) and were unchanged after adjusting for confounders and multiple comparison correction. Interestingly, no significant association was found for C16:0, C22:0, C24:0, C26:0, and C22:1 ceramide species, C24:0 dihydroceramide, or total ceramides. Our analysis reveals that specific long‐chain ceramides strongly associate with low cardiovascular fitness in older adults and may be implicated in the pathogenesis of low fitness with aging. Longitudinal studies are needed to further validate these associations and investigate the relationship between ceramides and health outcomes.
Collapse
Affiliation(s)
- Elisa Fabbri
- Longitudinal Studies Section Translational Gerontology Branch National Institute on Aging National Institutes of Health Baltimore MD 21224 USA
- Department of Medical and Surgical Sciences University of Bologna Bologna Italy
| | - An Yang
- Laboratory of Behavioral Neuroscience, Intramural Research Program National Institute on Aging National Institutes of Health Baltimore MD 21224 USA
| | - Eleanor M. Simonsick
- Longitudinal Studies Section Translational Gerontology Branch National Institute on Aging National Institutes of Health Baltimore MD 21224 USA
| | - Chee W. Chia
- Longitudinal Studies Section Translational Gerontology Branch National Institute on Aging National Institutes of Health Baltimore MD 21224 USA
| | - Marco Zoli
- Department of Medical and Surgical Sciences University of Bologna Bologna Italy
| | - Norman J. Haughey
- Department of Neurology Johns Hopkins University School of Medicine Baltimore MD 21224 USA
| | - Michelle M. Mielke
- Department of Health Science Research and Neurology Mayo Clinic Rochester MN 55905 USA
| | - Luigi Ferrucci
- Longitudinal Studies Section Translational Gerontology Branch National Institute on Aging National Institutes of Health Baltimore MD 21224 USA
| | - Paul M. Coen
- Translational Research Institute for Metabolism and Diabetes Florida Hospital Orlando FL 32804 USA
| |
Collapse
|
237
|
Chang X, Wang Z, Zhang J, Yan H, Bian H, Xia M, Lin H, Jiang J, Gao X. Lipid profiling of the therapeutic effects of berberine in patients with nonalcoholic fatty liver disease. J Transl Med 2016; 14:266. [PMID: 27629750 PMCID: PMC5024486 DOI: 10.1186/s12967-016-0982-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 07/18/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND We recently demonstrated a positive effect of berberine on nonalcoholic fatty liver disease patients after 16 weeks of treatment by comparing mere lifestyle intervention in type 2 diabetes patients with berberine treatment, which decreased the content of hepatic fat. However, the potential mechanisms of the clinical effects are unclear. We used a lipidomic approach to characterize the state of lipid metabolism as reflected in the circulation of subjects with nonalcoholic fatty liver disease (NAFLD) before and after berberine treatment. METHODS Liquid chromatography-mass spectrometry evaluated the various lipid metabolites in serum samples obtained from the participants (41 patients in the berberine group and 39 patients in the mere lifestyle intervention group) before and after treatment. RESULTS A total of 256 serum lipid molecular species were identified and quantified. Both treatments regulated various types of lipids in metabolic pathways, such as free fatty acids, phosphoglycerides and glycerides, in metabolic pathways, but berberine induced a substantially greater change in serum lipid species compared with mere lifestyle intervention after treatment. Berberine also caused obvious differences on ceramides. Berberine treatment markedly decreased serum levels of ceramide and ceramide-1-phosphate. CONCLUSIONS Berberine altered circulating ceramides, which may underlie the improvement in fatty liver disease. ClinicalTrials.gov NCT00633282, Registered March 3, 2008.
Collapse
Affiliation(s)
- Xinxia Chang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.,Institute of Metabolic Disease of Fudan University, Shanghai, China
| | - Zhe Wang
- Institute of Materia Medica, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, 100050, China
| | - Jinlan Zhang
- Institute of Materia Medica, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, 100050, China
| | - Hongmei Yan
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.,Institute of Metabolic Disease of Fudan University, Shanghai, China
| | - Hua Bian
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.,Institute of Metabolic Disease of Fudan University, Shanghai, China
| | - Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.,Institute of Metabolic Disease of Fudan University, Shanghai, China
| | - Huandong Lin
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.,Institute of Metabolic Disease of Fudan University, Shanghai, China
| | - Jiandong Jiang
- Institute of Materia Medica, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, 100050, China.
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China. .,Institute of Metabolic Disease of Fudan University, Shanghai, China.
| |
Collapse
|
238
|
Kurko J, Tringham M, Tanner L, Näntö-Salonen K, Vähä-Mäkilä M, Nygren H, Pöhö P, Lietzen N, Mattila I, Olkku A, Hyötyläinen T, Orešič M, Simell O, Niinikoski H, Mykkänen J. Imbalance of plasma amino acids, metabolites and lipids in patients with lysinuric protein intolerance (LPI). Metabolism 2016; 65:1361-75. [PMID: 27506743 DOI: 10.1016/j.metabol.2016.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 04/26/2016] [Accepted: 05/20/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Lysinuric protein intolerance (LPI [MIM 222700]) is an aminoaciduria with defective transport of cationic amino acids in epithelial cells in the small intestine and proximal kidney tubules due to mutations in the SLC7A7 gene. LPI is characterized by protein malnutrition, failure to thrive and hyperammonemia. Many patients also suffer from combined hyperlipidemia and chronic kidney disease (CKD) with an unknown etiology. METHODS Here, we studied the plasma metabolomes of the Finnish LPI patients (n=26) and healthy control individuals (n=19) using a targeted platform for analysis of amino acids as well as two analytical platforms with comprehensive coverage of molecular lipids and polar metabolites. RESULTS Our results demonstrated that LPI patients have a dichotomy of amino acid profiles, with both decreased essential and increased non-essential amino acids. Altered levels of metabolites participating in pathways such as sugar, energy, amino acid and lipid metabolism were observed. Furthermore, of these metabolites, myo-inositol, threonic acid, 2,5-furandicarboxylic acid, galactaric acid, 4-hydroxyphenylacetic acid, indole-3-acetic acid and beta-aminoisobutyric acid associated significantly (P<0.001) with the CKD status. Lipid analysis showed reduced levels of phosphatidylcholines and elevated levels of triacylglycerols, of which long-chain triacylglycerols associated (P<0.01) with CKD. CONCLUSIONS This study revealed an amino acid imbalance affecting the basic cellular metabolism, disturbances in plasma lipid composition suggesting hepatic steatosis and fibrosis and novel metabolites correlating with CKD in LPI. In addition, the CKD-associated metabolite profile along with increased nitrite plasma levels suggests that LPI may be characterized by increased oxidative stress and apoptosis, altered microbial metabolism in the intestine and uremic toxicity.
Collapse
Affiliation(s)
- Johanna Kurko
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland.
| | - Maaria Tringham
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland.
| | - Laura Tanner
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland; Department of Clinical Genetics, Turku University Hospital, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Kirsti Näntö-Salonen
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Mari Vähä-Mäkilä
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Heli Nygren
- VTT Technical Research Centre of Finland, Tietotie 2, P.O. Boxs 1000, Espoo 02044 VTT, Finland.
| | - Päivi Pöhö
- Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, P.O. Boxs 56, Helsinki 00014, Finland.
| | - Niina Lietzen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland.
| | - Ismo Mattila
- Steno Diabetes Center A/S, Niels Steensens Vej 2, 2820 Gentofte, Denmark.
| | - Anu Olkku
- Eastern Finland Laboratory Centre, Puijonlaaksontie 2, 70210 Kuopio, Finland.
| | - Tuulia Hyötyläinen
- Steno Diabetes Center A/S, Niels Steensens Vej 2, 2820 Gentofte, Denmark.
| | - Matej Orešič
- Steno Diabetes Center A/S, Niels Steensens Vej 2, 2820 Gentofte, Denmark.
| | - Olli Simell
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Harri Niinikoski
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Juha Mykkänen
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland; Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland.
| |
Collapse
|
239
|
Characterization of the role of sphingomyelin synthase 2 in glucose metabolism in whole-body and peripheral tissues in mice. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:688-702. [DOI: 10.1016/j.bbalip.2016.04.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 04/20/2016] [Accepted: 04/30/2016] [Indexed: 11/22/2022]
|
240
|
Aburasayn H, Al Batran R, Ussher JR. Targeting ceramide metabolism in obesity. Am J Physiol Endocrinol Metab 2016; 311:E423-35. [PMID: 27382035 DOI: 10.1152/ajpendo.00133.2016] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/04/2016] [Indexed: 12/12/2022]
Abstract
Obesity is a major health concern that increases the risk for insulin resistance, type 2 diabetes (T2D), and cardiovascular disease. Thus, an enormous research effort has been invested into understanding how obesity-associated dyslipidemia and obesity-induced alterations in lipid metabolism increase the risk for these diseases. Accordingly, it has been proposed that the accumulation of lipid metabolites in organs such as the liver, skeletal muscle, and heart is critical to these obesity-induced pathologies. Ceramide is one such lipid metabolite that accumulates in tissues in response to obesity, and both pharmacological and genetic strategies that reduce tissue ceramide levels yield salutary actions on overall metabolic health. We will review herein why ceramide accumulates in tissues during obesity and how an increase in intracellular ceramide impacts cellular signaling and function as well as potential mechanisms by which reducing intracellular ceramide levels improves insulin resistance, T2D, atherosclerosis, and heart failure. Because a reduction in skeletal muscle ceramide levels is frequently associated with improvements in insulin sensitivity in humans, the beneficial findings reported for reducing ceramides in preclinical studies may have clinical application in humans. Therefore, modulating ceramide metabolism may be a novel, exciting target for preventing and/or treating obesity-related diseases.
Collapse
Affiliation(s)
- Hanin Aburasayn
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; and Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Rami Al Batran
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; and Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; and Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
241
|
Petersen MC, Jurczak MJ. Rebuttal from Max C. Petersen and Michael J. Jurczak. J Physiol 2016; 594:3177-8. [PMID: 26997320 DOI: 10.1113/jp272137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 02/10/2016] [Indexed: 11/08/2022] Open
Affiliation(s)
- Max C Petersen
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Michael J Jurczak
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
242
|
Kusminski CM, Bickel PE, Scherer PE. Targeting adipose tissue in the treatment of obesity-associated diabetes. Nat Rev Drug Discov 2016; 15:639-660. [PMID: 27256476 DOI: 10.1038/nrd.2016.75] [Citation(s) in RCA: 490] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Adipose tissue regulates numerous physiological processes, and its dysfunction in obese humans is associated with disrupted metabolic homeostasis, insulin resistance and type 2 diabetes mellitus (T2DM). Although several US-approved treatments for obesity and T2DM exist, these are limited by adverse effects and a lack of effective long-term glucose control. In this Review, we provide an overview of the role of adipose tissue in metabolic homeostasis and assess emerging novel therapeutic strategies targeting adipose tissue, including adipokine-based strategies, promotion of white adipose tissue beiging as well as reduction of inflammation and fibrosis.
Collapse
Affiliation(s)
- Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center
| | - Perry E Bickel
- Division of Endocrinology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center
| |
Collapse
|
243
|
Scherer PE. The Multifaceted Roles of Adipose Tissue-Therapeutic Targets for Diabetes and Beyond: The 2015 Banting Lecture. Diabetes 2016; 65:1452-61. [PMID: 27222389 PMCID: PMC4878420 DOI: 10.2337/db16-0339] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 03/28/2016] [Indexed: 12/29/2022]
Abstract
The Banting Medal for Scientific Achievement is the highest scientific award of the American Diabetes Association (ADA). Given in memory of Sir Frederick Banting, one of the key investigators in the discovery of insulin, the Banting Medal is awarded annually for scientific excellence, recognizing significant long-term contributions to the understanding, treatment, or prevention of diabetes. Philipp E. Scherer, PhD, of the Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX, received the prestigious award at the ADA's 75th Scientific Sessions, 5-9 June 2015, in Boston, MA. He presented the Banting Lecture, "The Multifaceted Roles of Adipose Tissue-Therapeutic Targets for Diabetes and Beyond," on Sunday, 7 June 2015.A number of different cell types contribute to the cellular architecture of adipose tissue. Although the adipocyte is functionally making important contributions to systemic metabolic homeostatis, several additional cell types contribute a supportive role to bestow maximal flexibility on the tissue with respect to many biosynthetic and catabolic processes, depending on the metabolic state. These cells include vascular endothelial cells, a host of immune cells, and adipocyte precursor cells and fibroblasts. Combined, these cell types give rise to a tissue with remarkable flexibility with respect to expansion and contraction, while optimizing the ability of the tissue to act as an endocrine organ through the release of many protein factors, critically influencing systemic lipid homeostasis and biochemically contributing many metabolites. Using an example from each of these categories-adiponectin as a key adipokine, sphingolipids as critical mediators of insulin sensitivity, and uridine as an important metabolite contributed by the adipocyte to the systemic pool-I will discuss the emerging genesis of the adipocyte over the past 20 years from metabolic bystander to key driver of metabolic flexibility.
Collapse
Affiliation(s)
- Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
244
|
Stern JH, Rutkowski JM, Scherer PE. Adiponectin, Leptin, and Fatty Acids in the Maintenance of Metabolic Homeostasis through Adipose Tissue Crosstalk. Cell Metab 2016; 23:770-84. [PMID: 27166942 PMCID: PMC4864949 DOI: 10.1016/j.cmet.2016.04.011] [Citation(s) in RCA: 682] [Impact Index Per Article: 85.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metabolism research has made tremendous progress over the last several decades in establishing the adipocyte as a central rheostat in the regulation of systemic nutrient and energy homeostasis. Operating at multiple levels of control, the adipocyte communicates with organ systems to adjust gene expression, glucoregulatory hormone exocytosis, enzymatic reactions, and nutrient flux to equilibrate the metabolic demands of a positive or negative energy balance. The identification of these mechanisms has great potential to identify novel targets for the treatment of diabetes and related metabolic disorders. Herein, we review the central role of the adipocyte in the maintenance of metabolic homeostasis, highlighting three critical mediators: adiponectin, leptin, and fatty acids.
Collapse
Affiliation(s)
- Jennifer H Stern
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joseph M Rutkowski
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
245
|
Schuchman EH. Acid ceramidase and the treatment of ceramide diseases: The expanding role of enzyme replacement therapy. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1459-71. [PMID: 27155573 DOI: 10.1016/j.bbadis.2016.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/18/2016] [Accepted: 05/03/2016] [Indexed: 01/20/2023]
Abstract
Ceramides are a diverse group of sphingolipids that play important roles in many biological processes. Acid ceramidase (AC) is one key enzyme that regulates ceramide metabolism. Early research on AC focused on the fact that it is the enzyme deficient in the rare genetic disorder, Farber Lipogranulomatosis. Recent research has revealed that deficiency of the same enzyme is responsible for a rare form of spinal muscular atrophy associated with myoclonic epilepsy (SMA-PME). Due to their diverse role in biology, accumulation of ceramides also has been implicated in the pathobiology of many other common diseases, including infectious lung diseases, diabetes, cancers and others. This has revealed the potential of AC as a therapy for many of these diseases. This review will focus on the biology of AC and the potential role of this enzyme in the treatment of human disease.
Collapse
Affiliation(s)
- Edward H Schuchman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
246
|
Ding Y, Xian X, Holland WL, Tsai S, Herz J. Low-Density Lipoprotein Receptor-Related Protein-1 Protects Against Hepatic Insulin Resistance and Hepatic Steatosis. EBioMedicine 2016; 7:135-45. [PMID: 27322467 PMCID: PMC4913705 DOI: 10.1016/j.ebiom.2016.04.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 03/31/2016] [Accepted: 04/01/2016] [Indexed: 11/15/2022] Open
Abstract
Low-density lipoprotein receptor-related protein-1 (LRP1) is a multifunctional uptake receptor for chylomicron remnants in the liver. In vascular smooth muscle cells LRP1 controls reverse cholesterol transport through platelet-derived growth factor receptor β (PDGFR-β) trafficking and tyrosine kinase activity. Here we show that LRP1 regulates hepatic energy homeostasis by integrating insulin signaling with lipid uptake and secretion. Somatic inactivation of LRP1 in the liver (hLRP1KO) predisposes to diet-induced insulin resistance with dyslipidemia and non-alcoholic hepatic steatosis. On a high-fat diet, hLRP1KO mice develop a severe Metabolic Syndrome secondary to hepatic insulin resistance, reduced expression of insulin receptors on the hepatocyte surface and decreased glucose transporter 2 (GLUT2) translocation. While LRP1 is also required for efficient cell surface insulin receptor expression in the absence of exogenous lipids, this latent state of insulin resistance is unmasked by exposure to fatty acids. This further impairs insulin receptor trafficking and results in increased hepatic lipogenesis, impaired fatty acid oxidation and reduced very low density lipoprotein (VLDL) triglyceride secretion. Hepatic LRP1 deficiency in a mouse model (hLRP1KO) predisposes to diet-induced insulin resistance, dyslipidemia, and obesity. Insulin resistance in the hLRP1KO mouse results from reduced cell surface expression of insulin receptor (IR) and impaired translocation of glucose transporter 2 (GLUT2). Excess fatty acids in hLRP1KO mice shift hepatic fatty acid metabolism from an oxidative to a synthetic state, resulting in hepatic steatosis.
LRP1 is a multifunctional transmembrane receptor with essential functions in lipoprotein metabolism and subcellular receptor tyrosine kinase trafficking. A mouse model of hepatic LRP1 deficiency integrates the hallmark findings in Metabolic Syndrome - insulin resistance, dyslipidemia, and hepatic steatosis - with impaired glucose metabolism and altered hepatic fatty acid metabolism as a consequence of reduced insulin receptor trafficking and signaling. These findings underscore the central role of LRP1 in overall energy homeostasis, and specifically liver glucose and fatty acid metabolism.
Collapse
Affiliation(s)
- Yinyuan Ding
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA; Key Laboratory of Medical Electrophysiology, Ministry of Education of China, China; Institute of Cardiovascular Research, Sichuan Medical University, Luzhou 646000, China
| | - Xunde Xian
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| | - William L Holland
- Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shirling Tsai
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA; Dallas VA Medical Center, Dallas, TX 75216, USA
| | - Joachim Herz
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
247
|
Summers SA, Goodpaster BH. CrossTalk proposal: Intramyocellular ceramide accumulation does modulate insulin resistance. J Physiol 2016; 594:3167-70. [PMID: 26996141 DOI: 10.1113/jp271676] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/10/2016] [Indexed: 12/12/2022] Open
Affiliation(s)
- Scott A Summers
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Bret H Goodpaster
- Sanford-Burnham Prebys Medical Discovery Institute, Lake Nona, FL, USA
| |
Collapse
|
248
|
Wang ZV, Scherer PE. Adiponectin, the past two decades. J Mol Cell Biol 2016; 8:93-100. [PMID: 26993047 DOI: 10.1093/jmcb/mjw011] [Citation(s) in RCA: 389] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 12/25/2015] [Indexed: 12/22/2022] Open
Abstract
Adiponectin is an adipocyte-specific factor, first described in 1995. Over the past two decades, numerous studies have elucidated the physiological functions of adiponectin in obesity, diabetes, inflammation, atherosclerosis, and cardiovascular disease. Adiponectin, elicited through cognate receptors, suppresses glucose production in the liver and enhances fatty acid oxidation in skeletal muscle, which together contribute to a beneficial metabolic action in whole body energy homeostasis. Beyond its role in metabolism, adiponectin also protects cells from apoptosis and reduces inflammation in various cell types via receptor-dependent mechanisms. Adiponectin, as a fat-derived hormone, therefore fulfills a critical role as an important messenger to communicate between adipose tissue and other organs. A better understanding of adiponectin actions, including the pros and cons, will advance our insights into basic mechanisms of metabolism and inflammation, and potentially pave the way toward novel means of pharmacological intervention to address pathophysiological changes associated with diabetes, atherosclerosis, and cardiometabolic disease.
Collapse
Affiliation(s)
- Zhao V Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| |
Collapse
|
249
|
Kauhanen D, Sysi-Aho M, Koistinen KM, Laaksonen R, Sinisalo J, Ekroos K. Development and validation of a high-throughput LC-MS/MS assay for routine measurement of molecular ceramides. Anal Bioanal Chem 2016; 408:3475-83. [PMID: 26922344 DOI: 10.1007/s00216-016-9425-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/09/2016] [Accepted: 02/15/2016] [Indexed: 12/16/2022]
Abstract
Monitoring the levels of the ceramides (Cer) d18:1/16:0, Cer d18:1/18:0, Cer d18:1/24:0, and Cer d18:1/24:1 and ratios thereof in human plasma empowers the prediction of fatal outcome of coronary artery disease (CAD). We describe a validated liquid chromatography-tandem mass spectrometry (LC-MS/MS) methodology for clinical-scaled measurement of the four distinct ceramides. Rapid plasma precipitation was accomplished in 96-well format. Excellent extraction recoveries in the range of 98-109% were achieved for each ceramide. Addition of corresponding D7-labeled ceramide standards facilitated precise quantification of each plasma ceramide species utilizing a novel short 5-min LC-MS/MS method. Neither matrix interference nor carryover was observed. Robust intra- and inter-assay accuracy and precision <15% at five different concentrations were obtained. Linear calibration lines with regressions, R(2) > 0.99, were achieved for all analytes. Short-term bench top, long-term plasma, and extract stability demonstrated that the distinct ceramides were stable in the conditions evaluated. The validity of the methodology was demonstrated by determining the precise ceramide concentrations in a small CAD case-control study. Thus, our LC-MS/MS methodology features simple sample preparation and short analysis time for accurate quantification of Cer d18:1/16:0, Cer d18:1/18:0, Cer d18:1/24:0, and Cer d18:1/24:1, designed for routine analysis.
Collapse
Affiliation(s)
| | | | | | | | - Juha Sinisalo
- Heart and Lung Center, Helsinki University Hospital, 00029 HUS, Helsinki, Finland
| | - Kim Ekroos
- Zora Biosciences, Biologinkuja 1, 02150, Espoo, Finland.
| |
Collapse
|
250
|
Gosejacob D, Jäger PS, Vom Dorp K, Frejno M, Carstensen AC, Köhnke M, Degen J, Dörmann P, Hoch M. Ceramide Synthase 5 Is Essential to Maintain C16:0-Ceramide Pools and Contributes to the Development of Diet-induced Obesity. J Biol Chem 2016; 291:6989-7003. [PMID: 26853464 DOI: 10.1074/jbc.m115.691212] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Indexed: 01/22/2023] Open
Abstract
Ceramides are bioactive sphingolipids, which are composed of sphingoid bases carrying acyl chains of various lengths. Ceramides are synthesized by a family of six ceramide synthases (CerS) in mammals, which produce ceramides with differentN-linked acyl chains. Increased ceramide levels are known to contribute to the development of obesity and insulin resistance. Recently, it has been demonstrated that the ceramide acylation pattern is of particular importance for an organism to maintain energy homeostasis. However, which of theCerSfamily members are involved in this process is not yet completely known. Using newly developedCerS5knock-out mice, we show here thatCerS5is essential to maintain cellular C16:0sphingolipid pools in lung, spleen, muscle, liver, and white adipose tissue. Glycerophospholipid levels inCerS5-deficient mice were not altered. We found a strong impact of CerS5-dependent ceramide synthesis in white adipose tissue after high fat diet feeding. In skeletal muscle, liver, and spleen, C16:0-ceramide levels were altered independent of feeding conditions. The loss ofCerS5is associated with reduced weight gain and improved systemic health, including maintenance of glucose homeostasis and reduced white adipose tissue inflammation after high fat diet challenge. Our findings indicate that reduction of endogenous C16:0-ceramide by genetic inhibition ofCerS5is sufficient to ameliorate obesity and its comorbidities.
Collapse
Affiliation(s)
- Dominic Gosejacob
- From the LIMES-Institute, Program Unit Development, Genetics and Molecular Physiology, Molecular Developmental Biology, University of Bonn, Carl-Troll-Strasse 31 and
| | - Philipp S Jäger
- From the LIMES-Institute, Program Unit Development, Genetics and Molecular Physiology, Molecular Developmental Biology, University of Bonn, Carl-Troll-Strasse 31 and
| | - Katharina Vom Dorp
- IMBIO, Molecular Biotechnology, University of Bonn, Karlrobert-Kreiten-Str. 13, 53115 Bonn, Germany
| | - Martin Frejno
- From the LIMES-Institute, Program Unit Development, Genetics and Molecular Physiology, Molecular Developmental Biology, University of Bonn, Carl-Troll-Strasse 31 and
| | - Anne C Carstensen
- From the LIMES-Institute, Program Unit Development, Genetics and Molecular Physiology, Molecular Developmental Biology, University of Bonn, Carl-Troll-Strasse 31 and
| | - Monika Köhnke
- From the LIMES-Institute, Program Unit Development, Genetics and Molecular Physiology, Molecular Developmental Biology, University of Bonn, Carl-Troll-Strasse 31 and
| | - Joachim Degen
- From the LIMES-Institute, Program Unit Development, Genetics and Molecular Physiology, Molecular Developmental Biology, University of Bonn, Carl-Troll-Strasse 31 and
| | - Peter Dörmann
- IMBIO, Molecular Biotechnology, University of Bonn, Karlrobert-Kreiten-Str. 13, 53115 Bonn, Germany
| | - Michael Hoch
- From the LIMES-Institute, Program Unit Development, Genetics and Molecular Physiology, Molecular Developmental Biology, University of Bonn, Carl-Troll-Strasse 31 and
| |
Collapse
|