1
|
Zhu JY, Chen M, Mu WJ, Luo HY, Li Y, Li S, Yan LJ, Li RY, Yin MT, Li X, Chen HM, Guo L. Exercise-induced anti-obesity effects in male mice generated by a FOXO1-KLF10 reinforcing loop promoting adipose lipolysis. Nat Commun 2025; 16:3111. [PMID: 40169574 PMCID: PMC11961606 DOI: 10.1038/s41467-025-58467-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 03/21/2025] [Indexed: 04/03/2025] Open
Abstract
Exercise combats obesity and metabolic disorders, but the underlying mechanism is incompletely understood. KLF10, a transcription factor involved in various biological processes, has an undefined role in adipose tissue and obesity. Here, we show that exercise facilitates adipocyte-derived KLF10 expression via SIRT1/FOXO1 pathway. Adipocyte-specific knockout of KLF10 blunts exercise-promoted white adipose browning, energy expenditure, fat loss, glucose tolerance in diet-induced obese male mice. Conversely, adipocyte-specific transgenic expression of KLF10 in male mice enhanced the above metabolic profits induced by exercise. Mechanistically, KLF10 interacts with FOXO1 and facilitates the recruitment of KDM4A to form a ternary complex on the promoter regions of Pnpla2 and Lipe genes to promote these key lipolytic genes expression by demethylating H3K9me3 on their promoters, which facilitates lipolysis to defend against obesity in male mice. As a downstream effector responding to exercise, adipose KLF10 could act as a potential target in the fight against obesity.
Collapse
Affiliation(s)
- Jie-Ying Zhu
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Min Chen
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Wang-Jing Mu
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Hong-Yang Luo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Yang Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Shan Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Lin-Jing Yan
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Ruo-Ying Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Meng-Ting Yin
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Xin Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Hu-Min Chen
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Liang Guo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China.
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China.
- Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
2
|
Wu L, Huang F, Yang L, Yang L, Sun Z, Zhang J, Xia S, Zhao H, Ding Y, Bian D, Li K. Interplay of FXN expression and lipolysis in white adipocytes plays a critical role in insulin sensitivity in Friedreich's ataxia mouse model. Sci Rep 2024; 14:19876. [PMID: 39191875 DOI: 10.1038/s41598-024-71099-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024] Open
Abstract
Frataxin (FXN) is required for iron-sulfur cluster biogenesis, and its loss causes the early-onset neurodegenerative disease Friedreich ataxia (FRDA). Loss of FXN is a susceptibility factor in the development of diabetes, a common metabolic complication after myocardial hypertrophy in patients with FRDA. The underlying mechanism of FXN deficient-induced hyperglycemia in FRDA is, however, poorly understood. In this study, we confirmed that the FXN deficiency mouse model YG8R develops insulin resistance in elder individuals by disturbing lipid metabolic homeostasis in adipose tissues. Evaluation of lipolysis, lipogenesis, and fatty acid β-oxidation showed that lipolysis is most severely affected in white adipose tissues. Consistently, FXN deficiency significantly decreased expression of lipolytic genes encoding adipose triglyceride lipase (Atgl) and hormone-sensitive lipase (Hsl) resulting in adipocyte enlargement and inflammation. Lipolysis induction by fasting or cold exposure remarkably upregulated FXN expression, though FXN deficiency lessened the competency of lipolysis compared with the control or wild type mice. Moreover, we found that the impairment of lipolysis was present at a young age, a few months earlier than hyperglycemia and insulin resistance. Forskolin, an activator of lipolysis, or pioglitazone, an agonist of PPARγ, improved insulin sensitivity in FXN-deficient adipocytes or mice. We uncovered the interplay between FXN expression and lipolysis and found that impairment of lipolysis, particularly the white adipocytes, is an early event, likely, as a primary cause for insulin resistance in FRDA patients at later age.
Collapse
Affiliation(s)
- Lin Wu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Fei Huang
- Endocrinology Department, Yancheng First People's Hospital, Affiliated Hospital of Medical School, Nanjing University, Yancheng, 224000, People's Republic of China
| | - Lu Yang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Liu Yang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Zichen Sun
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Jinghua Zhang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Siyu Xia
- Endocrinology Department, Yancheng First People's Hospital, Affiliated Hospital of Medical School, Nanjing University, Yancheng, 224000, People's Republic of China
| | - Hongting Zhao
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Yibing Ding
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Dezhi Bian
- Endocrinology Department, Yancheng First People's Hospital, Affiliated Hospital of Medical School, Nanjing University, Yancheng, 224000, People's Republic of China.
| | - Kuanyu Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China.
| |
Collapse
|
3
|
Chen L, Wang J, Ren Y, Ma Y, Liu J, Jiang H, Liu C. Artesunate improves glucose and lipid metabolism in db/db mice by regulating the metabolic profile and the MAPK/PI3K/Akt signalling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155382. [PMID: 38382280 DOI: 10.1016/j.phymed.2024.155382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/23/2023] [Accepted: 01/20/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Diabetes is a metabolic disorder characterized by chronic hyperglycaemia. Chronic metabolic abnormalities and long-term hyperglycaemia may result in a wide range of acute and chronic consequences. Previous studies have demonstrated that artesunate(ART) has antidiabetic, anti-inflammatory, antiatherosclerotic, and other beneficial effects, but the specific regulatory mechanism is not completely clear. AIM This study investigated the effects of ART on metabolic disorders in type 2 diabetes mellitus (T2DM) model db/db mice and explored the underlying mechanisms involved. METHODS C57BL/KsJ-db/db mice were used to identify the targets and molecular mechanism of ART. Metabolomic methods were used to evaluate the efficacy of ART in improving T2DM-related metabolic disorders. Network pharmacology and transcriptomic sequencing were used to analyse the targets and pathways of ART in T2DM. Finally, molecular biology experiments were performed to verify the key targets and pathways selected by network pharmacology and transcriptomic analyses. RESULTS After a 7-week ART intervention (160 mg/kg), the glucose and lipid metabolism levels of the db/db mice improved. Additionally, the oxidative stress indices, namely, the MDA and SOD levels, significantly improved (p<0.01). Linoleic acid and glycerophospholipid metabolism, amino acid metabolism, bile acid synthesis, and purine metabolism disorders in db/db mice were partially corrected after ART treatment. Network pharmacology analysis identified important targets of ART for the treatment of metabolic disorders in T2DM . These targets are involved in key signalling pathways, including the highest scores observed for the PI3K/Akt signalling pathway. Transcriptomic analysis revealed that ART could activate the MAPK signalling pathway and two key gene targets, HGK and GADD45. Immunoblotting revealed that ART increases p-PI3K, p-AKT, Glut2, and IRS1 protein expression and suppresses the phosphorylation of p38, ERK1/2, and JNK, returning HGK and GADD45 to their preartesunate levels. CONCLUSION Treatment of db/db mice with 160 mg/kg ART for 7 weeks significantly reduced fasting blood glucose and lipid levels. It also improved metabolic imbalances in amino acids, lipids, purines, and bile acids, thereby improving metabolic disorders. These effects are achieved by activating the PI3K/AKT pathway and inhibiting the MAPK pathway, thus demonstrating the efficacy of the drug.
Collapse
Affiliation(s)
- Lulu Chen
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China; Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Jialin Wang
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Yanshuang Ren
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Yujin Ma
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Jie Liu
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Hongwei Jiang
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| | - Chuanxin Liu
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| |
Collapse
|
4
|
Engin A. Lipid Storage, Lipolysis, and Lipotoxicity in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:97-129. [PMID: 39287850 DOI: 10.1007/978-3-031-63657-8_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The ratio of free fatty acid (FFA) turnover decreases significantly with the expansion of white adipose tissue. Adipose tissue and dietary saturated fatty acid levels significantly correlate with an increase in fat cell size and number. The G0/G1 switch gene 2 increases lipid content in adipocytes and promotes adipocyte hypertrophy through the restriction of triglyceride (triacylglycerol: TAG) turnover. Hypoxia in obese adipose tissue due to hypertrophic adipocytes results in excess deposition of extracellular matrix (ECM) components. Cluster of differentiation (CD) 44, as the main receptor of the extracellular matrix component regulates cell-cell and cell-matrix interactions including diet-induced insulin resistance. Excess TAGs, sterols, and sterol esters are surrounded by the phospholipid monolayer surface and form lipid droplets (LDs). Once LDs are formed, they grow up because of the excessive amount of intracellular FFA stored and reach a final size. The ratio of FFA turnover/lipolysis decreases significantly with increases in the degree of obesity. Dysfunctional adipose tissue is unable to expand further to store excess dietary lipids, increased fluxes of plasma FFAs lead to ectopic fatty acid deposition and lipotoxicity. Reduced neo-adipogenesis and dysfunctional lipid-overloaded adipocytes are hallmarks of hypertrophic obesity linked to insulin resistance. Obesity-associated adipocyte death exhibits feature of necrosis-like programmed cell death. Adipocyte death is a prerequisite for the transition from hypertrophic to hyperplastic obesity. Increased adipocyte number in obesity has life-long effects on white adipose tissue mass. The positive correlation between the adipose tissue volume and magnetic resonance imaging proton density fat fraction estimation is used for characterization of the obesity phenotype, as well as the risk stratification and selection of appropriate treatment strategies. In obese patients with type 2 diabetes, visceral adipocytes exposed to chronic/intermittent hyperglycemia develop a new microRNAs' (miRNAs') expression pattern. Visceral preadipocytes memorize the effect of hyperglycemia via changes in miRNAs' expression profile and contribute to the progression of diabetic phenotype. Nonsteroidal anti-inflammatory drugs, metformin, and statins can be beneficial in treating the local or systemic consequences of white adipose tissue inflammation. Rapamycin inhibits leptin-induced LD formation. Collectively, in this chapter, the concept of adipose tissue remodeling in response to adipocyte death or adipogenesis, and the complexity of LD interactions with the other cellular organelles are reviewed. Furthermore, clinical perspective of fat cell turnover in obesity is also debated.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
5
|
Cremer J, Brohée L, Dupont L, Lefevre C, Peiffer R, Saarinen AM, Peulen O, Bindels L, Liu J, Colige A, Deroanne CF. Acidosis-induced regulation of adipocyte G0S2 promotes crosstalk between adipocytes and breast cancer cells as well as tumor progression. Cancer Lett 2023:216306. [PMID: 37442366 DOI: 10.1016/j.canlet.2023.216306] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/07/2023] [Accepted: 07/07/2023] [Indexed: 07/15/2023]
Abstract
Bidirectional interactions between cancer cells and their microenvironment govern tumor progression. Among the stromal cells in this microenvironment, adipocytes have been reported to upregulate cancer cell migration and invasion by producing fatty acids. Conversely, cancer cells alter adipocyte phenotype notably via increased lipolysis. We aimed to identify the mechanisms through which cancer cells trigger adipocyte lipolysis and evaluate the functional consequences on cancer progression. Here, we show that cancer cell-induced acidification of the extracellular medium strongly promotes preadipocyte lipolysis through a mechanism that does not involve lipophagy but requires adipose triglyceride lipase (ATGL) activity. This increased lipolysis is triggered mainly by attenuation of the G0/G1 switch gene 2 (G0S2)-induced inhibition of ATGL. G0S2-mediated regulation in preadipocytes affects their communication with breast cancer cells, modifying the phenotype of the cancer cells and increasing their resistance to chemotherapeutic agents in vitro. Furthermore, we demonstrate that the adipocyte-specific overexpression of G0S2 impairs mammary tumor growth and lung metastasis formation in vivo. Our results highlight the importance of acidosis in cancer cell-adipocyte crosstalk and identify G0S2 as the main regulator of cancer-induced lipolysis, regulating tumor establishment and spreading.
Collapse
Affiliation(s)
- Julie Cremer
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Laura Brohée
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Laura Dupont
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Camille Lefevre
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Avenue Mounier 73, B1.73.11, 1200, Brussels, Belgium
| | - Raphaël Peiffer
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Alicia M Saarinen
- Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona Scottsdale, AZ, USA
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Laure Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Avenue Mounier 73, B1.73.11, 1200, Brussels, Belgium
| | - Jun Liu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Alain Colige
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Christophe F Deroanne
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, Avenue Hippocrate 13, 4000, Liège, Belgium.
| |
Collapse
|
6
|
Bridge-Comer PE, Reilly SM. Measuring the Rate of Lipolysis in Ex vivo Murine Adipose Tissue and Primary Preadipocytes Differentiated In Vitro. J Vis Exp 2023:10.3791/65106. [PMID: 37010285 PMCID: PMC10583296 DOI: 10.3791/65106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
Abstract
Adipocytes store energy in the form of triglycerides in lipid droplets. This energy can be mobilized via lipolysis, where the fatty acid side chains are sequentially cleaved from the glycerol backbone, resulting in the release of free fatty acids and glycerol. Due to the low expression of glycerol kinase in white adipocytes, glycerol re-uptake rates are negligible, while fatty acid re-uptake is dictated by the fatty acid binding capacity of media components such as albumin. Both glycerol and fatty acid release into media can be quantified by colorimetric assays to determine the lipolytic rate. By measuring these factors at multiple time points, one can determine the linear rate of lipolysis with high confidence. Here, we provide a detailed protocol for the measurement of lipolysis in in vitro differentiated adipocytes and ex vivo adipose tissue from mice. This protocol may also be optimized for other preadipocyte cell lines or adipose tissue from other organisms; considerations and optimization parameters are discussed. This protocol is designed to be useful in determining and comparing the rate of adipocyte lipolysis between mouse models and treatments.
Collapse
Affiliation(s)
- Pania E Bridge-Comer
- Weill Center for Metabolic Health, Department of Medicine, Weill Cornell Medicine
| | - Shannon M Reilly
- Weill Center for Metabolic Health, Department of Medicine, Weill Cornell Medicine;
| |
Collapse
|
7
|
Wei X, Li L, Zhao J, Huo Y, Hu X, Lu J, Pi J, Zhang W, Xu L, Yao Y, Xu J. BAP31 depletion inhibited adipogenesis, repressed lipolysis and promoted lipid droplets abnormal growth via attenuating Perilipin1 proteasomal degradation. Int J Biol Sci 2023; 19:1713-1730. [PMID: 37063427 PMCID: PMC10092757 DOI: 10.7150/ijbs.82178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/25/2023] [Indexed: 04/18/2023] Open
Abstract
BAP31 expression was robustly decreased in obese white adipose tissue (WAT). To investigate the roles of BAP31 in lipid metabolism, adipocyte-specific conditional knockout mice (BAP31-ASKO) were generated. BAP31-ASKO mice grow normally as controls, but exhibited reduced lipid accumulation in WAT. Histomorphometric analysis reported increased adipocyte size in BAP31-ASKO mice. Mouse embryonic fibroblasts (MEFs) were induced to differentiation to adipocytes, showed reduced induction of adipogenic markers and attenuated adipogenesis in BAP31-deficient MEFs. BAP31-deficiency inhibited fasting-induced PKA signaling activation and the fasting response. β3-adrenergic receptor agonist-induced lipolysis also was reduced, accompanied by reduced free-fatty acids and glycerol release, and impaired agonist-induced lipolysis from primary adipocytes and adipose explants. BAP31 interacts with Perilipin1 via C-terminal cytoplasmic portion on lipid droplets (LDs) surface. Depletion of BAP31 repressed Perilipin1 proteasomal degradation, enhanced Perilipin1 expression and blocked LDs degradation, which promoted LDs abnormal growth and supersized LDs formation, resulted in adipocyte expansion, thus impaired insulin signaling and aggravated pro-inflammation in WAT. BAP31-deficiency increased phosphatidylcholine/phosphatidylethanolamine ratio, long chain triglycerides and most phospholipids contents. Overall, BAP31-deficiency inhibited adipogenesis and lipid accumulation in WAT, decreased LDs degradation and promoted LDs abnormal growth, pointing the critical roles in modulating LDs dynamics and homeostasis via proteasomal degradation system in adipocytes.
Collapse
Affiliation(s)
- Xueying Wei
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, Liaoning, China
| | - Liya Li
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, Liaoning, China
| | - Jie Zhao
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, Liaoning, China
| | - Yan Huo
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, Liaoning, China
| | - Xiaodi Hu
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, Liaoning, China
| | - Jingyi Lu
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, Liaoning, China
| | - Jingbo Pi
- School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Wei Zhang
- Department of Hepatobiliary Surgery, General Hospital of Northern Theater Command of the Chinese People's Liberation Army, Shenyang, 110016, Liaoning, China
| | - Lisheng Xu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, 110819, Liaoning, China
| | - Yudong Yao
- Department of Electrical and Computer Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Jialin Xu
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, Liaoning, China
- ✉ Corresponding author: Jialin Xu, Ph. D., Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, Liaoning, China Phone: (+86) 2483656117, E-mail:
| |
Collapse
|
8
|
Campbell LE, Anderson AM, Chen Y, Johnson SM, McMahon CE, Liu J. Identification of motifs and mechanisms for lipid droplet targeting of the lipolytic inhibitors G0S2 and HIG2. J Cell Sci 2022; 135:285951. [PMID: 36420951 PMCID: PMC10112975 DOI: 10.1242/jcs.260236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 11/15/2022] [Indexed: 11/27/2022] Open
Abstract
G0S2 and HIG2 are two selective inhibitors of ATGL (also known as PNPLA2), the key enzyme for intracellular lipolysis. Whereas G0S2 regulates triglyceride (TG) mobilization in adipocytes and hepatocytes, HIG2 functions to enhance intracellular TG accumulation under hypoxic conditions. A homologous hydrophobic domain (HD) is shared by G0S2 and HIG2 (also known as HILPDA) for binding to ATGL. However, the determinants of their lipid droplet (LD) localization are unknown. Here, we study how G0S2 and HIG2 are targeted to LDs, and identify both ATGL-independent and -dependent mechanisms. Structural prediction and studies in cells reveal that ATGL-independent localization of G0S2 to both the endoplasmic reticulum (ER) and LDs is mediated by a hairpin structure consisting of two hydrophobic sequences. Positively charged residues in the hinge region play a crucial role in sorting G0S2, which initially localizes to ER, to LDs. Interestingly, the role of these positive charges becomes dispensable when ATGL is co-expressed. In comparison, HIG2, which lacks a similar hairpin structure, is dependent on ATGL for its full LD targeting. Thus, our studies identify specific structural features and mechanisms for mediating accumulation of these two ATGL inhibitors on LDs.
Collapse
Affiliation(s)
- Latoya E Campbell
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine & Science, Rochester, MN 55905, USA.,College of Health Solutions, Arizona State University, Tempe, AZ 85281, USA
| | - Aaron M Anderson
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine & Science, Rochester, MN 55905, USA.,Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Yongbin Chen
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine & Science, Rochester, MN 55905, USA
| | - Scott M Johnson
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine & Science, Rochester, MN 55905, USA.,Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Cailin E McMahon
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine & Science, Rochester, MN 55905, USA
| | - Jun Liu
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine & Science, Rochester, MN 55905, USA.,Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic in Rochester, Rochester, MN 55905, USA
| |
Collapse
|
9
|
Middle-Aged Lpaatδ-Deficient Mice Have Altered Metabolic Measures. Life (Basel) 2022; 12:life12111717. [DOI: 10.3390/life12111717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/19/2022] [Accepted: 10/23/2022] [Indexed: 11/16/2022] Open
Abstract
Lysophosphatidic acid acyltransferases/acylglycerophosphate acyltransferases (LPAATs/AGPATs) are a group of homologous enzymes that catalyze the formation of phosphatidic acid (PA) from lysophosphatidic acid. We have previously reported that LPAATδ/AGPAT4 localizes to mitochondria, suggesting a potential role in energy metabolism. However, in prior studies of young Lpaatδ-deficient mice (age 9–12 weeks old), we found no differences in body weights, food intakes, activity levels, respiratory gas exchange, or energy expenditure compared to their wildtype (Wt) littermates. To test whether Lpaatδ−/− mice may develop differences in metabolic measures with advancing age, we recorded body weights and food intakes, and used metabolic chambers to assess ambulatory and locomotor activity levels, oxygen consumption (VO2), carbon dioxide production (VCO2), respiratory exchange ratio (RER), and total energy expenditure (heat). Fourteen-month-old Lpaatδ−/− mice had significantly lower mean body weights compared to Wt littermate controls (44.6 ± 1.08 g vs. 53.5 ± 0.42 g, respectively), but no significant differences in food intake or activity levels. This phenotypic difference was accompanied by significantly elevated 24 h daily, and 12 h light and dark photoperiod average VO2 (~20% higher) and VCO2 (~30% higher) measures, as well as higher RER and total energy expenditure (heat) values compared to Wt control littermates. Thus, an age-related metabolic phenotype is evident in Lpaatδ−/− mice. Future studies should examine the role of the lipid-modifying enzyme LPAATδ across the lifespan for greater insight into its role in normal and pathophysiology.
Collapse
|
10
|
Abstract
Metabolic disorders related to obesity are largely dependent on adipose tissue hypertrophy, which involves adipocyte hypertrophy and increased adipogenesis. Adiposize is regulated by lipid accumulation as a result of increased lipogenesis (mainly lipid uptake in mature adipocytes) and reduced lipolysis. Using realtime 2D cell culture analyses of lipid uptake, we show (1) that high glucose concentration (4.5 g/L) was required to accumulate oleic acid increasing lipid droplet size until unilocularization similar to mature adipocytes in few days, (2) oleic acid reduced Peroxisome-Proliferator Activated Receptor Gamma (PPARG) gene transcription and (3) insulin counteracted oleic acid-induced increase of lipid droplet size. Although the lipolytic activity observed in high versus low glucose (1 g/L) conditions was not altered, insulin was found to inhibit oleic acid induced gene transcription required for lipid storage such as Cell Death Inducing DFFA Like Effectors (CIDEC) and G0S2 (G0 switch gene S2), possibly through PPARA activity. Although this signalling pathway requires more detailed investigation, the results point out the differential mechanisms involved in the pro-adipogenic effect of insulin in absence versus its protective effect on adiposity in presence of oleic acid uptake. Abbreviations: AICAR, 5-Aminoimidazole-4-carboxamide-1-D-ribofuranoside; AMPK, AMP-Activated protein kinase, ASCs, adipose stem cell; ATGL, adipose triglyceride lipase; BSA, Bovine serum albumin; CEBPA, CCAAT enhancer binding protein alpha; CIDEs, Cell Death Inducing DFFA Like Effectors; dA, differentiated adipocyte; DMEM, Dulbecco’s Modified Eagle’s Medium; FABPs, Fatty Acid Binding Proteins; FAT/CD36, Fatty acid translocase; FCS, Foetal calf serum; FN1, fibronectin 1; FFA, free fatty acid; G0S2, G0 switch gene S2; GLUTs, Glucose transporters; GPR120, G protein-coupled receptor 120; HG, high glucose; HSL, hormone sensitive lipase; INSR, insulin receptor; LG, low glucose; OA, oleic acid; PBS, Phosphate buffer saline; PPARs, Peroxisome-Proliferator Activated Receptors; PKA, Protein kinase cyclic AMP-dependent; PKG, Protein kinase cyclic GMP dependent; PTGS2, cytochrome oxidase 2; RTCA, realtime cell analysis; TG, triglyceride.
Collapse
Affiliation(s)
- Emmanuelle Berger
- University of Lyon, UMR Ecologie Microbienne Lyon (LEM), Research Team "Bacterial Opportunistic Pathogens and Environment" (BPOE), CNRS 5557, INRAE 1418, Université Claude Bernard Lyon 1, VetAgro Sup, 69622 Villeurbanne ou 69363 Lyon, France
| | - Alain Géloën
- University of Lyon, UMR Ecologie Microbienne Lyon (LEM), Research Team "Bacterial Opportunistic Pathogens and Environment" (BPOE), CNRS 5557, INRAE 1418, Université Claude Bernard Lyon 1, VetAgro Sup, 69622 Villeurbanne ou 69363 Lyon, France
| |
Collapse
|
11
|
Riegler-Berket L, Wechselberger L, Cerk IK, Padmanabha Das KM, Viertlmayr R, Kulminskaya N, Rodriguez Gamez CF, Schweiger M, Zechner R, Zimmermann R, Oberer M. Residues of the minimal sequence of G0S2 collectively contribute to ATGL inhibition while C-and N-terminal extensions promote binding to ATGL. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159105. [PMID: 35026402 DOI: 10.1016/j.bbalip.2021.159105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/29/2021] [Accepted: 12/17/2021] [Indexed: 11/25/2022]
Abstract
The protein encoded by the G0/G1 switch gene 2 (G0S2) is a potent inhibitor of adipose triglyceride lipase (ATGL) and thus an important regulator of intracellular lipolysis. Since dysfunction of lipolysis is associated with metabolic diseases including diabetes and obesity, inhibition of ATGL is considered a therapeutic strategy. G0S2 interacts with ATGL's patatin-domain to mediate non-competitive inhibition, however atomic details of the inhibition mechanism are incompletely understood. Sequences of G0S2 from higher organisms show a highly conserved N-terminal part, including a hydrophobic region covering amino acids 27 to 42. We show that predicted G0S2 orthologs from platypus, chicken and Japanese rice-fish are able to inhibit human and mouse ATGL, emphasizing the contribution of conserved amino acid to ATGL inhibition. Our site directed mutagenesis and truncation studies give insights in the protein-protein interaction on a per-residue level. We determine that the minimal sequence required for ATGL inhibition ranges from amino acids 20 to 44. Residues Y27, V28, G30, A34 G37, V39 or L42 within this sequence play a substantial role in ATGL inhibition. Furthermore, we show that unspecific interactions of the N-terminal part (amino acids 20-27) of the minimal sequence facilitate the interaction to ATGL. Our studies also demonstrate that full-length G0S2 shows higher tolerance to specific single amino acid exchanges in the hydrophobic region due to the stronger contributions of unspecific interactions. However, exchanges of more than one amino-acid in the hydrophobic region also result in the loss of function as ATGL inhibitor even in the full-length protein.
Collapse
Affiliation(s)
- L Riegler-Berket
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - L Wechselberger
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - I K Cerk
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - K M Padmanabha Das
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - R Viertlmayr
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - N Kulminskaya
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | | | - M Schweiger
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; BioTechMed Graz, 8010 Graz, Austria
| | - R Zechner
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; BioTechMed Graz, 8010 Graz, Austria; BioHealth Field of Excellence, University of Graz, 8010 Graz, Austria
| | - R Zimmermann
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; BioTechMed Graz, 8010 Graz, Austria; BioHealth Field of Excellence, University of Graz, 8010 Graz, Austria
| | - M Oberer
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; BioTechMed Graz, 8010 Graz, Austria; BioHealth Field of Excellence, University of Graz, 8010 Graz, Austria.
| |
Collapse
|
12
|
Lipid droplets associated perilipins protein insights into finding a therapeutic target approach to cure non-alcoholic fatty liver disease (NAFLD). FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2022. [DOI: 10.1186/s43094-021-00395-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Abstract
Background
Non-alcoholic fatty liver disease (NAFLD) is now the most common form of chronic liver disease in the world, and it’s linked to a slew of other risk factors including diabetes, obesity, dysbiosis and inflammatory bowel disease. More than 30 years ago, a patient was diagnosed with fatty liver with excessive fat accumulation in hepatocytes, a disorder known as hepatosteatosis. There will be no promising therapeutic medicines available from 1980 to 2021 which can reverse the fatty liver to normal liver state. In this review, we highlighted on lipid droplet associated protein which play a major role in accumulation of fat in liver cells and how these cellular pathway could be a promising therapeutic approach to treat the fatty liver disease.
Main body
Over the last few decades, Western countries follow a high-fat diet and change their lifestyle pattern due to certain metabolic disorders prevalence rate is very high all over the world. NAFLD is a major health issue and burden globally nowadays. Researchers are trying to find out the potential therapeutic target to combat the disease. The exact pathophysiology of the disease is still unclear. In the present decades. There is no Food and Drug Administration approved drugs are available to reverse the chronic condition of the disease. Based on literature survey, lipid droplets and their associated protein like perilipins play an eminent role in body fat regulation. In this review, we explain all types of perilipins such as perilipin1-5 (PLIN1-5) and their role in the pathogenesis of fatty liver which will be helpful to find the novel pharmacological target to treat the fatty liver.
Conclusion
In this review, majorly focussed on how fat is get deposited into hepatocytes follow the cellular signalling involved during lipid droplet biogenesis and leads to NAFLD. However, up to date still there mechanism of action is unclear. In this review, we hypothesized that lipid droplets associated proteins like perilipins could be better pharmacological target to reverse the chronic stage of fatty liver disease and how these lipid droplets associated proteins hide a clue to maintain the normal lipid homeostasis in the human body.
Collapse
|
13
|
Grabner GF, Xie H, Schweiger M, Zechner R. Lipolysis: cellular mechanisms for lipid mobilization from fat stores. Nat Metab 2021; 3:1445-1465. [PMID: 34799702 DOI: 10.1038/s42255-021-00493-6] [Citation(s) in RCA: 367] [Impact Index Per Article: 91.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/15/2021] [Indexed: 12/13/2022]
Abstract
The perception that intracellular lipolysis is a straightforward process that releases fatty acids from fat stores in adipose tissue to generate energy has experienced major revisions over the last two decades. The discovery of new lipolytic enzymes and coregulators, the demonstration that lipophagy and lysosomal lipolysis contribute to the degradation of cellular lipid stores and the characterization of numerous factors and signalling pathways that regulate lipid hydrolysis on transcriptional and post-transcriptional levels have revolutionized our understanding of lipolysis. In this review, we focus on the mechanisms that facilitate intracellular fatty-acid mobilization, drawing on canonical and noncanonical enzymatic pathways. We summarize how intracellular lipolysis affects lipid-mediated signalling, metabolic regulation and energy homeostasis in multiple organs. Finally, we examine how these processes affect pathogenesis and how lipolysis may be targeted to potentially prevent or treat various diseases.
Collapse
Affiliation(s)
- Gernot F Grabner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Hao Xie
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
14
|
Povero D, Johnson SM, Liu J. Hypoxia, hypoxia-inducible gene 2 (HIG2)/HILPDA, and intracellular lipolysis in cancer. Cancer Lett 2020; 493:71-79. [PMID: 32818550 PMCID: PMC11218043 DOI: 10.1016/j.canlet.2020.06.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/27/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022]
Abstract
Tumor tissues are chronically exposed to hypoxia owing to aberrant vascularity. Hypoxia induces metabolic alterations in cancer, thereby promoting aggressive malignancy and metastasis. While previous efforts largely focused on adaptive responses in glucose and glutamine metabolism, recent studies have begun to yield important insight into the hypoxic regulation of lipid metabolic reprogramming in cancer. Emerging evidence points to lipid droplet (LD) accumulation as a hallmark of hypoxic cancer cells. One critical underlying mechanism involves the inhibition of adipose triglyceride lipase (ATGL)-mediated intracellular lipolysis by a small protein encoded by hypoxia-inducible gene 2 (HIG2), also known as hypoxia inducible lipid droplet associated (HILPDA). In this review we summarize and discuss recent key findings on hypoxia-dependent regulation of metabolic adaptations especially lipolysis in cancer. We also pose several questions and hypotheses pertaining to the metabolic impact of lipolytic regulation in cancer under hypoxia and during hypoxia-reoxygenation transition.
Collapse
Affiliation(s)
- Davide Povero
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Division of Endocrinology, Rochester, MN, 55905, USA
| | - Scott M Johnson
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Mayo Clinic College of Medicine & Science, Rochester, MN, 55905, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, 55905, USA
| | - Jun Liu
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Division of Endocrinology, Rochester, MN, 55905, USA.
| |
Collapse
|
15
|
Murakami M, Sato H, Taketomi Y. Updating Phospholipase A 2 Biology. Biomolecules 2020; 10:E1457. [PMID: 33086624 PMCID: PMC7603386 DOI: 10.3390/biom10101457] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/09/2020] [Accepted: 10/15/2020] [Indexed: 12/30/2022] Open
Abstract
The phospholipase A2 (PLA2) superfamily contains more than 50 enzymes in mammals that are subdivided into several distinct families on a structural and biochemical basis. In principle, PLA2 has the capacity to hydrolyze the sn-2 position of glycerophospholipids to release fatty acids and lysophospholipids, yet several enzymes in this superfamily catalyze other reactions rather than or in addition to the PLA2 reaction. PLA2 enzymes play crucial roles in not only the production of lipid mediators, but also membrane remodeling, bioenergetics, and body surface barrier, thereby participating in a number of biological events. Accordingly, disturbance of PLA2-regulated lipid metabolism is often associated with various diseases. This review updates the current state of understanding of the classification, enzymatic properties, and biological functions of various enzymes belonging to the PLA2 superfamily, focusing particularly on the novel roles of PLA2s in vivo.
Collapse
Affiliation(s)
- Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan; (H.S.); (Y.T.)
| | | | | |
Collapse
|
16
|
Inazumi T, Yamada K, Shirata N, Sato H, Taketomi Y, Morita K, Hohjoh H, Tsuchiya S, Oniki K, Watanabe T, Sasaki Y, Oike Y, Ogata Y, Saruwatari J, Murakami M, Sugimoto Y. Prostaglandin E2-EP4 Axis Promotes Lipolysis and Fibrosis in Adipose Tissue Leading to Ectopic Fat Deposition and Insulin Resistance. Cell Rep 2020; 33:108265. [DOI: 10.1016/j.celrep.2020.108265] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 09/10/2020] [Accepted: 09/22/2020] [Indexed: 12/24/2022] Open
|
17
|
Lee J, Kim DH, Lee K. Current Approaches and Applications in Avian Genome Editing. Int J Mol Sci 2020; 21:ijms21113937. [PMID: 32486292 PMCID: PMC7312999 DOI: 10.3390/ijms21113937] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 01/02/2023] Open
Abstract
Advances in genome-editing technologies and sequencing of animal genomes enable researchers to generate genome-edited (GE) livestock as valuable animal models that benefit biological researches and biomedical and agricultural industries. As birds are an important species in biology and agriculture, their genome editing has gained significant interest and is mainly performed by using a primordial germ cell (PGC)-mediated method because pronuclear injection is not practical in the avian species. In this method, PGCs can be isolated, cultured, genetically edited in vitro, and injected into a recipient embryo to produce GE offspring. Recently, a couple of GE quail have been generated by using the newly developed adenovirus-mediated method. Without technically required in vitro procedures of the PGC-mediated method, direct injection of adenovirus into the avian blastoderm in the freshly laid eggs resulted in the production of germ-line chimera and GE offspring. As more approaches are available in avian genome editing, avian research in various fields will progress rapidly. In this review, we describe the development of avian genome editing and scientific and industrial applications of GE avian species.
Collapse
Affiliation(s)
- Joonbum Lee
- Department of Animal Sciences, The Ohio State University, Columbus, OH 43210, USA; (J.L.); (D.-H.K.)
- The Ohio State University Interdisciplinary Human Nutrition Program, The Ohio State University, Columbus, OH 43210, USA
| | - Dong-Hwan Kim
- Department of Animal Sciences, The Ohio State University, Columbus, OH 43210, USA; (J.L.); (D.-H.K.)
| | - Kichoon Lee
- Department of Animal Sciences, The Ohio State University, Columbus, OH 43210, USA; (J.L.); (D.-H.K.)
- The Ohio State University Interdisciplinary Human Nutrition Program, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: ; Tel.: +1-614-688-7963
| |
Collapse
|
18
|
Miladinovic D, Cusick T, Mahon KL, Haynes AM, Cortie CH, Meyer BJ, Stricker PD, Wittert GA, Butler LM, Horvath LG, Hoy AJ. Assessment of Periprostatic and Subcutaneous Adipose Tissue Lipolysis and Adipocyte Size from Men with Localized Prostate Cancer. Cancers (Basel) 2020; 12:cancers12061385. [PMID: 32481537 PMCID: PMC7352157 DOI: 10.3390/cancers12061385] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/21/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
The prostate is surrounded by periprostatic adipose tissue (PPAT), the thickness of which has been associated with more aggressive prostate cancer (PCa). There are limited data regarding the functional characteristics of PPAT, how it compares to subcutaneous adipose tissue (SAT), and whether in a setting of localized PCa, these traits are altered by obesity or disease aggressiveness. PPAT and SAT were collected from 60 men (age: 42–78 years, BMI: 21.3–35.6 kg/m2) undergoing total prostatectomy for PCa. Compared to SAT, adipocytes in PPAT were smaller, had the same basal rates of fatty acid release (lipolysis) yet released less polyunsaturated fatty acid species, and were more sensitive to isoproterenol-stimulated lipolysis. Basal lipolysis of PPAT was increased in men diagnosed with less aggressive PCa (Gleason score (GS) ≤ 3 + 4) compared to men with more aggressive PCa (GS ≥ 4 + 3) but no other measured adipocyte parameters related to PCa aggressiveness. Likewise, there was no difference in PPAT lipid biology between lean and obese men. In conclusion, lipid biological features of PPAT do differ from SAT; however, we did not observe any meaningful difference in ex vivo PPAT biology that is associated with PCa aggressiveness or obesity. As such, our findings do not support a relationship between altered PCa behavior in obese men and the metabolic reprogramming of PPAT.
Collapse
Affiliation(s)
- Dushan Miladinovic
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales 2006, Australia;
| | - Thomas Cusick
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, New South Wales 2010, Australia; (T.C.); (K.L.M.); (A.-M.H.); (P.D.S.); (L.G.H.)
| | - Kate L. Mahon
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, New South Wales 2010, Australia; (T.C.); (K.L.M.); (A.-M.H.); (P.D.S.); (L.G.H.)
- Discipline of Medicine, Central Clinical School, The University of Sydney School of Medicine, Faculty of Medicine and Health, The University of Sydney, New South Wales 2006, Australia
- Department of Medical Oncology, Chris O’Brien Lifehouse, New South Wales 2050, Australia
- Royal Prince Alfred Hospital, New South Wales 2050, Australia
| | - Anne-Maree Haynes
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, New South Wales 2010, Australia; (T.C.); (K.L.M.); (A.-M.H.); (P.D.S.); (L.G.H.)
| | - Colin H. Cortie
- School of Medicine, Lipid Research Centre, Molecular Horizons, University of Wollongong, New South Wales 2522, Australia; (C.H.C.); (B.J.M.)
- Illawarra Medical Research Institute, University of Wollongong, New South Wales 2522, Australia
| | - Barbara J. Meyer
- School of Medicine, Lipid Research Centre, Molecular Horizons, University of Wollongong, New South Wales 2522, Australia; (C.H.C.); (B.J.M.)
- Illawarra Medical Research Institute, University of Wollongong, New South Wales 2522, Australia
| | - Phillip D. Stricker
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, New South Wales 2010, Australia; (T.C.); (K.L.M.); (A.-M.H.); (P.D.S.); (L.G.H.)
- St. Vincent’s Clinical School, The University of New South Wales, New South Wales 2010, Australia
- St. Vincent’s Prostate Cancer Centre, St. Vincent’s Clinic, New South Wales 2010, Australia
| | - Gary A. Wittert
- South Australian Health and Medical Research Institute, South Australia 5000, Australia; (G.A.W.); (L.M.B.)
- School of Medicine and Freemasons Foundation Centre for Men’s Health, University of Adelaide, South Australia 5000, Australia
| | - Lisa M. Butler
- South Australian Health and Medical Research Institute, South Australia 5000, Australia; (G.A.W.); (L.M.B.)
- School of Medicine and Freemasons Foundation Centre for Men’s Health, University of Adelaide, South Australia 5000, Australia
| | - Lisa G. Horvath
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, New South Wales 2010, Australia; (T.C.); (K.L.M.); (A.-M.H.); (P.D.S.); (L.G.H.)
- Discipline of Medicine, Central Clinical School, The University of Sydney School of Medicine, Faculty of Medicine and Health, The University of Sydney, New South Wales 2006, Australia
- Department of Medical Oncology, Chris O’Brien Lifehouse, New South Wales 2050, Australia
- Royal Prince Alfred Hospital, New South Wales 2050, Australia
| | - Andrew J. Hoy
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales 2006, Australia;
- Correspondence:
| |
Collapse
|
19
|
Yvan-Charvet L, Ivanov S. Metabolic Reprogramming of Macrophages in Atherosclerosis: Is It All about Cholesterol? J Lipid Atheroscler 2020; 9:231-242. [PMID: 32821733 PMCID: PMC7379089 DOI: 10.12997/jla.2020.9.2.231] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/16/2020] [Accepted: 02/11/2020] [Indexed: 12/20/2022] Open
Abstract
Hypercholesterolemia contributes to the chronic inflammatory response during the progression of atherosclerosis, in part by favoring cholesterol loading in macrophages and other immune cells. However, macrophages encounter a substantial amount of other lipids and nutrients after ingesting atherogenic lipoprotein particles or clearing apoptotic cells, increasing their metabolic load and impacting their behavior during atherosclerosis plaque progression. This review examines whether and how fatty acids and glucose shape the cellular metabolic reprogramming of macrophages in atherosclerosis to modulate the onset phase of inflammation and the later resolution stage, in which the balance is tipped toward tissue repair.
Collapse
Affiliation(s)
- Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Stoyan Ivanov
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| |
Collapse
|
20
|
Hofer P, Taschler U, Schreiber R, Kotzbeck P, Schoiswohl G. The Lipolysome-A Highly Complex and Dynamic Protein Network Orchestrating Cytoplasmic Triacylglycerol Degradation. Metabolites 2020; 10:E147. [PMID: 32290093 PMCID: PMC7240967 DOI: 10.3390/metabo10040147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/03/2020] [Accepted: 04/08/2020] [Indexed: 12/25/2022] Open
Abstract
The catabolism of intracellular triacylglycerols (TAGs) involves the activity of cytoplasmic and lysosomal enzymes. Cytoplasmic TAG hydrolysis, commonly termed lipolysis, is catalyzed by the sequential action of three major hydrolases, namely adipose triglyceride lipase, hormone-sensitive lipase, and monoacylglycerol lipase. All three enzymes interact with numerous protein binding partners that modulate their activity, cellular localization, or stability. Deficiencies of these auxiliary proteins can lead to derangements in neutral lipid metabolism and energy homeostasis. In this review, we summarize the composition and the dynamics of the complex lipolytic machinery we like to call "lipolysome".
Collapse
Affiliation(s)
- Peter Hofer
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; (P.H.); (U.T.); (R.S.)
| | - Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; (P.H.); (U.T.); (R.S.)
| | - Renate Schreiber
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; (P.H.); (U.T.); (R.S.)
| | - Petra Kotzbeck
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria;
| | - Gabriele Schoiswohl
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; (P.H.); (U.T.); (R.S.)
| |
Collapse
|
21
|
Karczewska-Kupczewska M, Nikołajuk A, Majewski R, Filarski R, Stefanowicz M, Matulewicz N, Strączkowski M. Changes in adipose tissue lipolysis gene expression and insulin sensitivity after weight loss. Endocr Connect 2020; 9:90-100. [PMID: 31905163 PMCID: PMC6993275 DOI: 10.1530/ec-19-0507] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 12/22/2019] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Insulin resistance is a major pathophysiological link between obesity and its metabolic complications. Weight loss (WL) is an effective tool to prevent obesity-related diseases; however, the mechanisms of an improvement in insulin sensitivity (IS) after weight-reducing interventions are not completely understood. The aim of the present study was to analyze the relationships between IS and adipose tissue (AT) expression of the genes involved in the regulation of lipolysis in obese subjects after WL. METHODS Fifty-two obese subjects underwent weight-reducing dietary intervention program. The control group comprised 20 normal-weight subjects, examined at baseline only. Hyperinsulinemic-euglycemic clamp and s.c. AT biopsy with subsequent gene expression analysis were performed before and after the program. RESULTS AT expression of genes encoding lipases (PNPLA2, LIPE and MGLL) and lipid-droplet proteins enhancing (ABHD5) and inhibiting lipolysis (PLIN1 and CIDEA) were decreased in obese individuals in comparison with normal-weight individuals. The group of 38 obese participants completed dietary intervention program and clamp studies, which resulted in a significant WL and an improvement in mean IS. However, in nine subjects from this group IS did not improve in response to WL. AT expression of PNPLA2, LIPE and PLIN1 increased only in the group without IS improvement. CONCLUSIONS Excessive lipolysis may prevent an improvement in IS during WL. The change in AT PNPLA2 and LIPE expression was a negative predictor of the change in IS after WL.
Collapse
Affiliation(s)
- Monika Karczewska-Kupczewska
- Department of Internal Medicine and Metabolic Diseases, Medical University of Białystok, Białystok, Poland
- Correspondence should be addressed to M Karczewska-Kupczewska:
| | - Agnieszka Nikołajuk
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research Polish Academy of Sciences, Olsztyn, Poland
| | - Radosław Majewski
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research Polish Academy of Sciences, Olsztyn, Poland
| | - Remigiusz Filarski
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research Polish Academy of Sciences, Olsztyn, Poland
| | - Magdalena Stefanowicz
- Department of Metabolic Diseases, Medical University of Białystok, Białystok, Poland
| | - Natalia Matulewicz
- Department of Metabolic Diseases, Medical University of Białystok, Białystok, Poland
| | - Marek Strączkowski
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
22
|
Kioka H, Kato H, Fujita T, Asano Y, Shintani Y, Yamazaki S, Tsukamoto O, Imamura H, Kogo M, Kitakaze M, Sakata Y, Takashima S. In vivo real-time ATP imaging in zebrafish hearts reveals G0s2 induces ischemic tolerance. FASEB J 2020; 34:2041-2054. [PMID: 31916304 DOI: 10.1096/fj.201901686r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/19/2019] [Accepted: 11/06/2019] [Indexed: 12/20/2022]
Abstract
Most eukaryotic cells generate adenosine triphosphate (ATP) through the oxidative phosphorylation system (OXPHOS) to support cellular activities. In cultured cell-based experiments, we recently identified the hypoxia-inducible protein G0/G1 switch gene 2 (G0s2) as a positive regulator of OXPHOS, and showed that G0s2 protects cultured cardiomyocytes from hypoxia. In this study, we examined the in vivo protective role of G0s2 against hypoxia by generating both loss-of-function and gain-of-function models of g0s2 in zebrafish. Zebrafish harboring transcription activator-like effector nuclease (TALEN)-mediated knockout of g0s2 lost hypoxic tolerance. Conversely, cardiomyocyte-specific transgenic zebrafish hearts exhibited strong tolerance against hypoxia. To clarify the mechanism by which G0s2 protects cardiac function under hypoxia, we introduced a mitochondrially targeted FRET-based ATP biosensor into zebrafish heart to visualize ATP dynamics in in vivo beating hearts. In addition, we employed a mosaic overexpression model of g0s2 to compare the contraction and ATP dynamics between g0s2-expressing and non-expressing cardiomyocytes, side-by-side within the same heart. These techniques revealed that g0s2-expressing cardiomyocyte populations exhibited preserved contractility coupled with maintained intra-mitochondrial ATP concentrations even under hypoxic condition. Collectively, these results demonstrate that G0s2 provides ischemic tolerance in vivo by maintaining ATP production, and therefore represents a promising therapeutic target for hypoxia-related diseases.
Collapse
Affiliation(s)
- Hidetaka Kioka
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hisakazu Kato
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Frontier Bioscience, Suita, Japan
| | - Takeshi Fujita
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan.,First Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Suita, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yasunori Shintani
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Satoru Yamazaki
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Osamu Tsukamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Frontier Bioscience, Suita, Japan
| | - Hiromi Imamura
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Mikihiko Kogo
- First Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Suita, Japan
| | - Masafumi Kitakaze
- Department of Clinical Research and Development, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Frontier Bioscience, Suita, Japan.,Japan Science and Technology Agency-Core Research for Evolutional Science and Technology (CREST), Kawaguchi, Japan
| |
Collapse
|
23
|
Di Giorgio E, Paluvai H, Picco R, Brancolini C. Genetic Programs Driving Oncogenic Transformation: Lessons from in Vitro Models. Int J Mol Sci 2019; 20:ijms20246283. [PMID: 31842516 PMCID: PMC6940909 DOI: 10.3390/ijms20246283] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022] Open
Abstract
Cancer complexity relies on the intracellular pleiotropy of oncogenes/tumor suppressors and in the strong interplay between tumors and micro- and macro-environments. Here we followed a reductionist approach, by analyzing the transcriptional adaptations induced by three oncogenes (RAS, MYC, and HDAC4) in an isogenic transformation process. Common pathways, in place of common genes became dysregulated. From our analysis it emerges that, during the process of transformation, tumor cells cultured in vitro prime some signaling pathways suitable for coping with the blood supply restriction, metabolic adaptations, infiltration of immune cells, and for acquiring the morphological plasticity needed during the metastatic phase. Finally, we identified two signatures of genes commonly regulated by the three oncogenes that successfully predict the outcome of patients affected by different cancer types. These results emphasize that, in spite of the heterogeneous mutational burden among different cancers and even within the same tumor, some common hubs do exist. Their location, at the intersection of the various signaling pathways, makes a therapeutic approach exploitable.
Collapse
|
24
|
Cho E, Kwon YJ, Ye DJ, Baek HS, Kwon TU, Choi HK, Chun YJ. G0/G1 Switch 2 Induces Cell Survival and Metastasis through Integrin-Mediated Signal Transduction in Human Invasive Breast Cancer Cells. Biomol Ther (Seoul) 2019; 27:591-602. [PMID: 31272137 PMCID: PMC6824625 DOI: 10.4062/biomolther.2019.063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/24/2019] [Accepted: 06/04/2019] [Indexed: 12/25/2022] Open
Abstract
Human breast cancer cell line, MDA-MB-231, is highly invasive and aggressive, compared to less invasive cell line, MCF-7. To explore the genes that might influence the malignancy of MDA-MB-231, DNA microarray analysis was performed. The results showed that G0/G1 switch 2 (G0S2) was one of the most highly expressed genes among the genes upregulated in MDA-MB-231. Although G0S2 acts as a direct inhibitor of adipose triglyceride lipase, action of G0S2 in cancer progression is not yet understood. To investigate whether G0S2 affects invasiveness of MDA-MB-231 cells, G0S2 expression was inhibited using siRNA, which led to decreased cell proliferation, migration, and invasion of MDA-MB-231 cells. Consequently, G0S2 inhibition inactivated integrinregulated FAK-Src signaling, which promoted Hippo signaling and inactivated ERK1/2 signaling. In addition, G0S2 downregulation decreased β-catenin expression, while E-cadherin expression was increased. It was demonstrated for the first time that G0S2 mediates the Hippo pathway and induces epithelial to mesenchymal transition (EMT). Taken together, our results suggest that G0S2 is a major factor contributing to cell survival and metastasis of MDA-MB-231 cells.
Collapse
Affiliation(s)
- Eunah Cho
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul 06974,
Republic of Korea
| | - Yeo-Jung Kwon
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul 06974,
Republic of Korea
| | - Dong-Jin Ye
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul 06974,
Republic of Korea
| | - Hyoung-Seok Baek
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul 06974,
Republic of Korea
| | - Tae-Uk Kwon
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul 06974,
Republic of Korea
| | - Hyung-Kyoon Choi
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul 06974,
Republic of Korea
| | - Young-Jin Chun
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul 06974,
Republic of Korea
| |
Collapse
|
25
|
Bartels ED, Guo S, Kousholt BS, Larsen JR, Hasenkam JM, Burnett J, Nielsen LB, Ashina M, Goetze JP. High doses of ANP and BNP exacerbate lipolysis in humans and the lipolytic effect of BNP is associated with cardiac triglyceride content in pigs. Peptides 2019; 112:43-47. [PMID: 30508635 DOI: 10.1016/j.peptides.2018.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022]
Abstract
Drugs facilitating the cardioprotective effects of natriuretic peptides are introduced in heart failure treatment. ANP and BNP also stimulate lipolysis and increase circulating concentrations of free fatty acids (FFAs); an aspect, however, thought to be confined to primates. We examined the lipolytic effect of natriuretic peptide infusion in healthy young men and evaluated the effect in a porcine model of myocardial ischemia and reperfusion. Six young healthy normotensive men underwent infusion with ANP, BNP, or CNP for 20 min. Blood samples were collected before, during, and after infusion for measurement of FFAs. In a porcine model of myocardial ischemia and reperfusion, animals were infused for 3 h with either BNP (n = 7) or saline (n = 5). Blood samples were collected throughout the infusion period, and cardiac tissue was obtained after infusion for lipid analysis. In humans, ANP infusion dose-dependently increased the FFA concentration in plasma 2.5-10-fold (baseline vs. 0.05 μg/kg/min P < 0.002) and with BNP 1.6-3.5-fold (P = 0.001, baseline vs. 0.02 μg/kg/min) 30 min after initiation of infusion. Infusion of CNP did not affect plasma FFA. In pigs, BNP infusion induced a 3.5-fold increase in plasma FFA (P < 0.0001), which remained elevated throughout the infusion period. Triglyceride content in porcine right cardiac ventricle tissue increased ∼5.5 fold in animals infused with BNP (P = 0.02). Natriuretic peptide infusion has similar lipolytic activity in human and pig. Our data suggest that short-term infusion increases the cardiac lipid content, and that the pig is a suitable model for studies of long-term effects mediated by natriuretic peptides.
Collapse
Affiliation(s)
- Emil D Bartels
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Song Guo
- Department of Neurology and Danish Headache Center, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte S Kousholt
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Jens R Larsen
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - J Michael Hasenkam
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - John Burnett
- Department of Cardiorenal physiology (Mayo Clinic, Rochester, MN, USA
| | - Lars B Nielsen
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Copenhagen University, Denmark; Aarhus University, Denmark
| | - Messoud Ashina
- Department of Neurology and Danish Headache Center, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Copenhagen University, Denmark
| | - Jens P Goetze
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark; Department of Cardiorenal physiology (Mayo Clinic, Rochester, MN, USA; Copenhagen University, Denmark
| |
Collapse
|
26
|
Ali SB, Turner JJO, Fountain SJ. Constitutive P2Y 2 receptor activity regulates basal lipolysis in human adipocytes. J Cell Sci 2018; 131:jcs.221994. [PMID: 30333139 DOI: 10.1242/jcs.221994] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/05/2018] [Indexed: 12/28/2022] Open
Abstract
White adipocytes are key regulators of metabolic homeostasis, which release stored energy as free fatty acids via lipolysis. Adipocytes possess both basal and stimulated lipolytic capacity, but limited information exists regarding the molecular mechanisms that regulate basal lipolysis. Here, we describe a mechanism whereby autocrine purinergic signalling and constitutive P2Y2 receptor activation suppresses basal lipolysis in primary human in vitro-differentiated adipocytes. We found that human adipocytes possess cytoplasmic Ca2+ tone due to ATP secretion and constitutive P2Y2 receptor activation. Pharmacological antagonism or knockdown of P2Y2 receptors increases intracellular cAMP levels and enhances basal lipolysis. P2Y2 receptor antagonism works synergistically with phosphodiesterase inhibitors in elevating basal lipolysis, but is dependent upon adenylate cyclase activity. Mechanistically, we suggest that the increased Ca2+ tone exerts an anti-lipolytic effect by suppression of Ca2+-sensitive adenylate cyclase isoforms. We also observed that acute enhancement of basal lipolysis following P2Y2 receptor antagonism alters the profile of secreted adipokines leading to longer-term adaptive decreases in basal lipolysis. Our findings demonstrate that basal lipolysis and adipokine secretion are controlled by autocrine purinergic signalling in human adipocytes.
Collapse
Affiliation(s)
- Seema B Ali
- Biomedical Research Centre, School of Biological Sciences, University of East Anglia, NR4 7TJ Norwich, UK
| | - Jeremy J O Turner
- Norfolk and Norwich University Hospital, Colney Lane, NR4 7TJ Norwich, UK
| | - Samuel J Fountain
- Biomedical Research Centre, School of Biological Sciences, University of East Anglia, NR4 7TJ Norwich, UK
| |
Collapse
|
27
|
Metabolite and lipoprotein responses and prediction of weight gain during breast cancer treatment. Br J Cancer 2018; 119:1144-1154. [PMID: 30401977 PMCID: PMC6220113 DOI: 10.1038/s41416-018-0211-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 07/06/2018] [Accepted: 07/10/2018] [Indexed: 12/18/2022] Open
Abstract
Background Breast cancer treatment has metabolic side effects, potentially affecting risk of cardiovascular disease (CVD) and recurrence. We aimed to compare alterations in serum metabolites and lipoproteins during treatment between recipients and non-recipients of chemotherapy, and describe metabolite profiles associated with treatment-related weight gain. Methods This pilot study includes 60 stage I/II breast cancer patients who underwent surgery and were treated according to national guidelines. Serum sampled pre-surgery and after 6 and 12 months was analysed by MR spectroscopy and mass spectrometry. In all, 170 metabolites and 105 lipoprotein subfractions were quantified. Results The metabolite and lipoprotein profiles of chemotherapy recipients and non-recipients changed significantly 6 months after surgery (p < 0.001). Kynurenine, the lipid signal at 1.55–1.60 ppm, ADMA, 2 phosphatidylcholines (PC aa C38:3, PC ae C42:1), alpha-aminoadipic acid, hexoses and sphingolipids were increased in chemotherapy recipients after 6 months. VLDL and small dense LDL increased after 6 months, while HDL decreased, with triglyceride enrichment in HDL and LDL. At baseline, weight gainers had less acylcarnitines, phosphatidylcholines, lyso-phosphatidylcholines and sphingolipids, and showed an inflammatory lipid profile. Conclusion Chemotherapy recipients exhibit metabolic changes associated with inflammation, altered immune response and increased risk of CVD. Altered lipid metabolism may predispose for treatment-related weight gain.
Collapse
|
28
|
Park TS, Park J, Lee JH, Park JW, Park BC. Disruption of G 0/G 1 switch gene 2 ( G0S2) reduced abdominal fat deposition and altered fatty acid composition in chicken. FASEB J 2018; 33:1188-1198. [PMID: 30085885 DOI: 10.1096/fj.201800784r] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Chicken as a food source is one of the most widespread domestic animals, and it has been used extensively as a research model. The clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) system is the most efficient and reliable tool for precise genome-targeted modification and has generated considerable excitement for industrial applications, as well as biologic science. Unlike in mammals, germline-transmittable primordial germ cells (PGCs) in chicken were used as an alternative strategy for the production of genetically altered chickens. Here, by combining the CRISPR-Cas9 platform and germ cell-mediated germline transmission, we generated G0/G1 switch gene 2 ( G0S2) knockout (KO) chickens, and G0S2 null KO chickens showed a dramatic reduction of abdominal fat deposition without affecting other economic traits. Additionally, G0S2 null KO chickens had altered fatty acid compositions in their blood and abdominal fat compared with wild-type chickens under normal dietary conditions. The global mRNA sequencing data showed that G0S2 disruption in chickens would activate the adipose tissue-specific peroxisomal oxidation pathway, and enoyl-coenzyme A (CoA), hydratase/3-hydroxyacyl CoA dehydrogenase might be a target molecule in metabolic homeostasis in the chicken adipose tissue. Our results demonstrate that the CRISPR-Cas9 system with chicken PGCs can facilitate the production of specific genome-edited chickens for practical applications, as well as basic research.-Park, T. S., Park, J., Lee, J. H., Park, J.-W., Park, B.-C. Disruption of G0/G1 switch gene 2 ( G0S2) reduced abdominal fat deposition and altered fatty acid composition in chicken.
Collapse
Affiliation(s)
- Tae Sub Park
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, Korea; and.,Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, South Korea
| | - Joonghoon Park
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, Korea; and.,Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, South Korea
| | - Jeong Hyo Lee
- Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, South Korea
| | - Jeong-Woong Park
- Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, South Korea
| | - Byung-Chul Park
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, Korea; and.,Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, South Korea
| |
Collapse
|
29
|
Matoba K, Lu Y, Zhang R, Chen ER, Sangwung P, Wang B, Prosdocimo DA, Jain MK. Adipose KLF15 Controls Lipid Handling to Adapt to Nutrient Availability. Cell Rep 2018; 21:3129-3140. [PMID: 29241541 DOI: 10.1016/j.celrep.2017.11.032] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 10/13/2017] [Accepted: 11/09/2017] [Indexed: 12/24/2022] Open
Abstract
Adipose tissue stores energy in the form of triglycerides. The ability to regulate triglyceride synthesis and breakdown based on nutrient status (e.g., fed versus fasted) is critical for physiological homeostasis and dysregulation of this process can contribute to metabolic disease. Whereas much is known about hormonal control of this cycle, transcriptional regulation is not well understood. Here, we show that the transcription factor Kruppel-like factor 15 (KLF15) is critical for the control of adipocyte lipid turnover. Mice lacking Klf15 in adipose tissue (AK15KO) display decreased adiposity and are protected from diet-induced obesity. Mechanistic studies suggest that adipose KLF15 regulates key genes of triglyceride synthesis and inhibits lipolytic action, thereby promoting lipid storage in an insulin-dependent manner. Finally, AK15KO mice demonstrate accelerated lipolysis and altered systemic energetics (e.g., locomotion, ketogenesis) during fasting conditions. Our study identifies adipose KLF15 as an essential regulator of adipocyte lipid metabolism and systemic energy balance.
Collapse
Affiliation(s)
- Keiichiro Matoba
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Yuan Lu
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Rongli Zhang
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Eric R Chen
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Panjamaporn Sangwung
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Benlian Wang
- Center for Proteomics and Bioinformatics and Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Domenick A Prosdocimo
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA.
| |
Collapse
|
30
|
Quiroga AD, Lehner R. Pharmacological intervention of liver triacylglycerol lipolysis: The good, the bad and the ugly. Biochem Pharmacol 2018; 155:233-241. [PMID: 30006193 DOI: 10.1016/j.bcp.2018.07.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/09/2018] [Indexed: 02/07/2023]
Abstract
Excessive triacylglycerol (TG) accumulation is the distinctive feature of obesity. In the liver, sustained TG accretion leads to nonalcoholic fatty liver disease (NAFLD), eventually progressing to non-alcoholic steatohepatitis (NASH) and cirrhosis, which is associated with complications including hepatic failure, hepatocellular carcinoma and death. Pharmacological interventions are actively pursued to prevent lipid accumulation in hepatocytes and, therefore, to ameliorate the associated pathophysiological conditions. Here, we sought to provide an overview of the pharmacological approaches to up- or downregulate the expression and activities of the enzymes involved in hepatic TG hydrolysis. Fatty acids (FA) released by hydrolysis of hepatic TG can be used for β-oxidation, signaling, and for very low-density lipoprotein (VLDL)-TG synthesis. Originally, lipolysis was believed to be centered in the adipose and to be catalyzed by only two lipases, hormone-sensitive lipase (HSL) and monoacylglycerol lipase (MAGL). However, genetic ablation of HSL expression in mice failed to erase TG hydrolysis in adipocytes leading to the identification of a third lipase termed adipose triglyceride lipase (ATGL). Although these three enzymes are considered to be the main players governing lipolysis in the adipocyte, other lipolytic enzymes have been described to contribute to hepatic TG metabolism. These include adiponutrin/patatin-like phospholipase domain containing 3 (PNPLA3), some members of the carboxylesterase family (CES/Ces), arylacetamide deacetylase (AADAC), lysosomal acid lipase (LAL) and hepatic lipase (HL). This review highlights the consequences of pharmacological interventions of liver lipases that degrade TG in cytosolic lipid droplets, in the endoplasmic reticulum, in the late endosomes/lysosomes and along the secretory route.
Collapse
Affiliation(s)
- Ariel D Quiroga
- Instituto de Fisiología Experimental (IFISE), Área Morfología, Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, UNR, Rosario, Argentina.
| | - Richard Lehner
- Group on Molecular and Cell Biology of Lipids, Department of Pediatrics, Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
31
|
Zechner R, Madeo F, Kratky D. Cytosolic lipolysis and lipophagy: two sides of the same coin. Nat Rev Mol Cell Biol 2017; 18:671-684. [PMID: 28852221 DOI: 10.1038/nrm.2017.76] [Citation(s) in RCA: 373] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fatty acids are the most efficient substrates for energy production in vertebrates and are essential components of the lipids that form biological membranes. Synthesis of triacylglycerols from non-esterified free fatty acids (FFAs) combined with triacylglycerol storage represents a highly efficient strategy to stockpile FFAs in cells and prevent FFA-induced lipotoxicity. Although essentially all vertebrate cells have some capacity to store and utilize triacylglycerols, white adipose tissue is by far the largest triacylglycerol depot and is uniquely able to supply FFAs to other tissues. The release of FFAs from triacylglycerols requires their enzymatic hydrolysis by a process called lipolysis. Recent discoveries thoroughly altered and extended our understanding of lipolysis. This Review discusses how cytosolic 'neutral' lipolysis and lipophagy, which utilizes 'acid' lipolysis in lysosomes, degrade cellular triacylglycerols as well as how these pathways communicate, how they affect lipid metabolism and energy homeostasis and how their dysfunction affects the pathogenesis of metabolic diseases. Answers to these questions will likely uncover novel strategies for the treatment of prevalent metabolic diseases.
Collapse
Affiliation(s)
- Rudolf Zechner
- BioTechMed-Graz, Mozartgasse 12, 8010 Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, 8010 Graz, Austria
| | - Frank Madeo
- BioTechMed-Graz, Mozartgasse 12, 8010 Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, 8010 Graz, Austria
| | - Dagmar Kratky
- BioTechMed-Graz, Mozartgasse 12, 8010 Graz, Austria
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| |
Collapse
|
32
|
Wang Y, Ma C, Sun Y, Li Y, Kang L, Jiang Y. Dynamic transcriptome and DNA methylome analyses on longissimus dorsi to identify genes underlying intramuscular fat content in pigs. BMC Genomics 2017; 18:780. [PMID: 29025412 PMCID: PMC5639760 DOI: 10.1186/s12864-017-4201-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 10/08/2017] [Indexed: 01/12/2023] Open
Abstract
Background The intramuscular fat content (IMF) refers to the amount of fat within muscles, including the sum of phospholipids mainly found in cell membranes, triglycerides and cholesterol, and is determined both by hyperplasia and hypertrophy of adipocyte during the development of pigs. The IMF content is an important economic trait that is genetically controlled by multiple genes. The Laiwu pig is an indigenous fatty pig breed distributed in North China, characterized by excessively higher level of IMF content (9%~12%), therefore, is suitable for the identification of genes controlling IMF variations. To identify genes underlying IMF deposition, we performed genome-wide transcriptome and methylome analyses on longissimus dorsi (LD) muscle in Laiwu pigs across four developmental stages. Results A total of 22,524 expressed genes were detected and 1158 differentially expressed genes (DEGs) were hierarchically clustered in the LD muscle over four developmental stages from 60 d to 400 d. These genes were significantly clustered into four temporal expression profiles, and genes participating in fat cell differentiation and lipid biosynthesis processes were identified. From 120 d to 240 d, the period with the maximum IMF deposition rate, the lipid biosynthesis related genes (FOSL1, FAM213B and G0S2), transcription factors (TFs) (EGR1, KLF5, SREBF2, TP53 and TWIST1) and enriched pathways (steroid biosynthesis and fatty acid biosynthesis) were revealed; and fat biosynthesis relevant genes showing differences in DNA methylation in gene body or intergenic region were detected, such as FASN, PVALB, ID2, SH3PXD2B and EGR1. Conclusions This study provides a comprehensive landscape of transcriptome of the LD muscle in Laiwu pigs ranging from 60 to 400 days old, and methylome of the LD muscle in 120 d and 240 d Laiwu pigs. A set of candidate genes and TFs involved in fat biosynthesis process were identified, which were probably responsible for IMF deposition. The results from this study would provide a reference for the identification of genes controlling IMF variation, and for exploring molecular mechanisms underlying IMF deposition in pigs. Electronic supplementary material The online version of this article (10.1186/s12864-017-4201-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuding Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, People's Republic of China.,Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, People's Republic of China
| | - Cai Ma
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, People's Republic of China
| | - Yi Sun
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, People's Republic of China
| | - Yi Li
- Central Hospital of Taian, Taian, 271018, People's Republic of China
| | - Li Kang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, People's Republic of China
| | - Yunliang Jiang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, People's Republic of China.
| |
Collapse
|
33
|
Engin A. Fat Cell and Fatty Acid Turnover in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:135-160. [PMID: 28585198 DOI: 10.1007/978-3-319-48382-5_6] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The ratio of free fatty acid (FFA) turnover decreases significantly with the expansion of white adipose tissue. Adipose tissue and dietary saturated fatty acid levels significantly correlate with an increase in fat cell size and number. Inhibition of adipose triglyceride lipase leads to an accumulation of triglyceride, whereas inhibition of hormone-sensitive lipase leads to the accumulation of diacylglycerol. The G0/G1 switch gene 2 increases lipid content in adipocytes and promotes adipocyte hypertrophy through the restriction of triglyceride turnover. Excess triacylglycerols (TAGs), sterols and sterol esters are surrounded by the phospholipid monolayer surface and form lipid droplets. Following the release of lipid droplets from endoplasmic reticulum, cytoplasmic lipid droplets increase their volume either by local TAG synthesis or by homotypic fusion. The number and the size of lipid droplet distribution is correlated with obesity. Obesity-associated adipocyte death exhibits feature of necrosis-like programmed cell death. NOD-like receptors family pyrin domain containing 3 (NLRP3) inflammasome-dependent caspase-1 activation in hypertrophic adipocytes induces obese adipocyte death by pyroptosis. Actually adipocyte death may be a prerequisite for the transition from hypertrophic to hyperplastic obesity. Major transcriptional factors, CCAAT/enhancer-binding proteins beta and delta, play a central role in the subsequent induction of critical regulators, peroxisome-proliferator-activated receptor gamma, CCAAT/enhancer-binding protein alpha and sterol regulatory element-binding protein 1, in the transcriptional control of adipogenesis in obesity.Collectively, in this chapter the concept of adipose tissue remodeling in response to adipocyte death or adipogenesis, and the complexity of lipid droplet interactions with the other cellular organelles are reviewed. Furthermore, in addition to lipid droplet growth, the functional link between the adipocyte-specific lipid droplet-associated protein and fatty acid turn-over is also debated.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey. .,, Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
34
|
Zhang X, Heckmann BL, Campbell LE, Liu J. G0S2: A small giant controller of lipolysis and adipose-liver fatty acid flux. Biochim Biophys Acta Mol Cell Biol Lipids 2017. [PMID: 28645852 DOI: 10.1016/j.bbalip.2017.06.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The discovery of adipose triglyceride lipase (ATGL) and its coactivator comparative gene identification-58 (CGI-58) provided a major paradigm shift in the understanding of intracellular lipolysis in both adipocytes and nonadipocyte cells. The subsequent discovery of G0/G1 switch gene 2 (G0S2) as a potent endogenous inhibitor of ATGL revealed a unique mechanism governing lipolysis and fatty acid (FA) availability. G0S2 is highly conserved in vertebrates, and exhibits cyclical expression pattern between adipose tissue and liver that is critical to lipid flux and energy homeostasis in these two tissues. Biochemical and cell biological studies have demonstrated that a direct interaction with ATGL mediates G0S2's inhibitory effects on lipolysis and lipid droplet degradation. In this review we examine evidence obtained from recent in vitro and in vivo studies that lends support to the proof-of-principle concept that G0S2 functions as a master regulator of tissue-specific balance of TG storage vs. mobilization, partitioning of metabolic fuels between adipose and liver, and the whole-body adaptive energy response. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, AZ, United States; HEAL(th) Program, Mayo Clinic, Scottsdale, AZ, United States
| | - Bradlee L Heckmann
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Latoya E Campbell
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Jun Liu
- Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, AZ, United States; HEAL(th) Program, Mayo Clinic, Scottsdale, AZ, United States; Division of Endocrinology, Mayo Clinic, Scottsdale, AZ, United States.
| |
Collapse
|
35
|
Mueller KM, Hartmann K, Kaltenecker D, Vettorazzi S, Bauer M, Mauser L, Amann S, Jall S, Fischer K, Esterbauer H, Müller TD, Tschöp MH, Magnes C, Haybaeck J, Scherer T, Bordag N, Tuckermann JP, Moriggl R. Adipocyte Glucocorticoid Receptor Deficiency Attenuates Aging- and HFD-Induced Obesity and Impairs the Feeding-Fasting Transition. Diabetes 2017; 66:272-286. [PMID: 27650854 DOI: 10.2337/db16-0381] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 09/14/2016] [Indexed: 11/13/2022]
Abstract
Glucocorticoids (GCs) are important regulators of systemic energy metabolism, and aberrant GC action is linked to metabolic dysfunctions. Yet, the extent to which normal and pathophysiological energy metabolism depend on the GC receptor (GR) in adipocytes remains unclear. Here, we demonstrate that adipocyte GR deficiency in mice significantly impacts systemic metabolism in different energetic states. Plasma metabolomics and biochemical analyses revealed a marked global effect of GR deficiency on systemic metabolite abundance and, thus, substrate partitioning in fed and fasted states. This correlated with a decreased lipolytic capacity of GR-deficient adipocytes under postabsorptive and fasting conditions, resulting from impaired signal transduction from β-adrenergic receptors to adenylate cyclase. Upon prolonged fasting, the impaired lipolytic response resulted in abnormal substrate utilization and lean mass wasting. Conversely, GR deficiency attenuated aging-/diet-associated obesity, adipocyte hypertrophy, and liver steatosis. Systemic glucose tolerance was improved in obese GR-deficient mice, which was associated with increased insulin signaling in muscle and adipose tissue. We conclude that the GR in adipocytes exerts central but diverging roles in the regulation of metabolic homeostasis depending on the energetic state. The adipocyte GR is indispensable for the feeding-fasting transition but also promotes adiposity and associated metabolic disorders in fat-fed and aged mice.
Collapse
Affiliation(s)
- Kristina M Mueller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Kerstin Hartmann
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | | | - Sabine Vettorazzi
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Mandy Bauer
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Lea Mauser
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Sabine Amann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Sigrid Jall
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH) and German Center for Diabetes Research (DZD), Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - Katrin Fischer
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH) and German Center for Diabetes Research (DZD), Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - Harald Esterbauer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH) and German Center for Diabetes Research (DZD), Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH) and German Center for Diabetes Research (DZD), Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - Christoph Magnes
- HEALTH Institute for Biomedicine and Health Sciences, JOANNEUM RESEARCH, Forschungsgesellschaft mbH, Graz, Austria
| | | | - Thomas Scherer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Natalie Bordag
- Center for Biomarker Research in Medicine, CBmed GmbH, Graz, Austria
| | - Jan P Tuckermann
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Medical University of Vienna, Vienna, Austria
| |
Collapse
|
36
|
Heckmann BL, Zhang X, Saarinen AM, Liu J. Regulation of G0/G1 Switch Gene 2 (G0S2) Protein Ubiquitination and Stability by Triglyceride Accumulation and ATGL Interaction. PLoS One 2016; 11:e0156742. [PMID: 27248498 PMCID: PMC4889065 DOI: 10.1371/journal.pone.0156742] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/18/2016] [Indexed: 12/31/2022] Open
Abstract
Intracellular triglyceride (TG) hydrolysis or lipolysis is catalyzed by the key intracellular triglyceride hydrolase, adipose triglyceride lipase (ATGL). The G0/G1 Switch Gene 2 (G0S2) was recently identified as the major selective inhibitor of ATGL and its hydrolase function. Since G0S2 levels are dynamically linked and rapidly responsive to nutrient status or metabolic requirements, the identification of its regulation at the protein level is of significant value. Earlier evidence from our laboratory demonstrated that G0S2 is a short-lived protein degraded through the proteasomal pathway. However, little is currently known regarding the underlying mechanisms. In the current study we find that 1) protein degradation is initiated by K48-linked polyubiquitination of the lysine- 25 in G0S2; and 2) G0S2 protein is stabilized in response to ATGL expression and TG accumulation. Mutation of lysine-25 of G0S2 abolished ubiquitination and increased protein stability. More importantly, G0S2 was stabilized via different mechanisms in the presence of ATGL vs. in response to fatty acid (FA)-induced TG accumulation. Furthermore, G0S2 protein but not mRNA levels were reduced in the adipose tissue of ATGL-deficient mice, corroborating the involvement of ATGL in the stabilization of G0S2. Taken together our data illustrate for the first time a crucial multifaceted mechanism for the stabilization of G0S2 at the protein level.
Collapse
Affiliation(s)
- Bradlee L. Heckmann
- Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, Arizona, United States of America
- Metabolic HEALth Program, Mayo Clinic, Scottsdale, Arizona, United States of America
- Mayo Graduate School, Rochester, Minnesota, United States of America
| | - Xiaodong Zhang
- Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, Arizona, United States of America
- Metabolic HEALth Program, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Alicia M. Saarinen
- Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, Arizona, United States of America
- Metabolic HEALth Program, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Jun Liu
- Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, Arizona, United States of America
- Metabolic HEALth Program, Mayo Clinic, Scottsdale, Arizona, United States of America
- * E-mail:
| |
Collapse
|
37
|
Laurens C, Badin PM, Louche K, Mairal A, Tavernier G, Marette A, Tremblay A, Weisnagel SJ, Joanisse DR, Langin D, Bourlier V, Moro C. G0/G1 Switch Gene 2 controls adipose triglyceride lipase activity and lipid metabolism in skeletal muscle. Mol Metab 2016; 5:527-537. [PMID: 27408777 PMCID: PMC4921782 DOI: 10.1016/j.molmet.2016.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Recent data suggest that adipose triglyceride lipase (ATGL) plays a key role in providing energy substrate from triglyceride pools and that alterations of its expression/activity relate to metabolic disturbances in skeletal muscle. Yet little is known about its regulation. We here investigated the role of the protein G0/G1 Switch Gene 2 (G0S2), recently described as an inhibitor of ATGL in white adipose tissue, in the regulation of lipolysis and oxidative metabolism in skeletal muscle. METHODS We first examined G0S2 protein expression in relation to metabolic status and muscle characteristics in humans. We next overexpressed and knocked down G0S2 in human primary myotubes to assess its impact on ATGL activity, lipid turnover and oxidative metabolism, and further knocked down G0S2 in vivo in mouse skeletal muscle. RESULTS G0S2 protein is increased in skeletal muscle of endurance-trained individuals and correlates with markers of oxidative capacity and lipid content. Recombinant G0S2 protein inhibits ATGL activity by about 40% in lysates of mouse and human skeletal muscle. G0S2 overexpression augments (+49%, p < 0.05) while G0S2 knockdown strongly reduces (-68%, p < 0.001) triglyceride content in human primary myotubes and mouse skeletal muscle. We further show that G0S2 controls lipolysis and fatty acid oxidation in a strictly ATGL-dependent manner. These metabolic adaptations mediated by G0S2 are paralleled by concomitant changes in glucose metabolism through the modulation of Pyruvate Dehydrogenase Kinase 4 (PDK4) expression (5.4 fold, p < 0.001). Importantly, downregulation of G0S2 in vivo in mouse skeletal muscle recapitulates changes in lipid metabolism observed in vitro. CONCLUSION Collectively, these data indicate that G0S2 plays a key role in the regulation of skeletal muscle ATGL activity, lipid content and oxidative metabolism.
Collapse
Affiliation(s)
- Claire Laurens
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - Pierre-Marie Badin
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - Katie Louche
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - Aline Mairal
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - Geneviève Tavernier
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - André Marette
- Department of Medicine, Canada; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada
| | - Angelo Tremblay
- Department of Kinesiology, Canada; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada
| | | | - Denis R Joanisse
- Department of Kinesiology, Canada; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada
| | - Dominique Langin
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France; Toulouse University Hospitals, Department of Clinical Biochemistry, Toulouse, France
| | - Virginie Bourlier
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - Cedric Moro
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France.
| |
Collapse
|
38
|
Yim CY, Sekula DJ, Hever-Jardine MP, Liu X, Warzecha JM, Tam J, Freemantle SJ, Dmitrovsky E, Spinella MJ. G0S2 Suppresses Oncogenic Transformation by Repressing a MYC-Regulated Transcriptional Program. Cancer Res 2016; 76:1204-13. [PMID: 26837760 DOI: 10.1158/0008-5472.can-15-2265] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 12/08/2015] [Indexed: 01/31/2023]
Abstract
Methylation-mediated silencing of G0-G1 switch gene 2 (G0S2) has been detected in a variety of solid tumors, whereas G0S2 induction is associated with remissions in patients with acute promyelocytic leukemia, implying that G0S2 may possess tumor suppressor activity. In this study, we clearly demonstrate that G0S2 opposes oncogene-induced transformation using G0s2-null immortalized mouse embryonic fibroblasts (MEF). G0s2-null MEFs were readily transformed with HRAS or EGFR treatment compared with wild-type MEFs. Importantly, restoration of G0S2 reversed HRAS-driven transformation. G0S2 is known to regulate fat metabolism by attenuating adipose triglyceride lipase (ATGL), but repression of oncogene-induced transformation by G0S2 was independent of ATGL inhibition. Gene expression analysis revealed an upregulation of gene signatures associated with transformation, proliferation, and MYC targets in G0s2-null MEFs. RNAi-mediated ablation and pharmacologic inhibition of MYC abrogated oncogene-induced transformation of G0s2-null MEFs. Furthermore, we found that G0S2 was highly expressed in normal breast tissues compared with malignant tissue. Intriguingly, high levels of G0S2 were also associated with a decrease in breast cancer recurrence rates, especially in estrogen receptor-positive subtypes, and overexpression of G0S2 repressed the proliferation of breast cancer cells in vitro. Taken together, these findings indicate that G0S2 functions as a tumor suppressor in part by opposing MYC activity, prompting further investigation of the mechanisms by which G0S2 silencing mediates MYC-induced oncogenesis in other malignancies.
Collapse
Affiliation(s)
- Christina Y Yim
- Department of Pharmacology and Toxicology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - David J Sekula
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mary P Hever-Jardine
- Department of Pharmacology and Toxicology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Xi Liu
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joshua M Warzecha
- Department of Pharmacology and Toxicology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Janice Tam
- Department of Pharmacology and Toxicology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Sarah J Freemantle
- Department of Pharmacology and Toxicology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Ethan Dmitrovsky
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael J Spinella
- Department of Pharmacology and Toxicology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire.
| |
Collapse
|
39
|
Skopp A, May M, Janke J, Kielstein H, Wunder R, Flade-Kuthe R, Kuthe A, Jordan J, Engeli S. Regulation of G0/G1 switch gene 2 (G0S2) expression in human adipose tissue. Arch Physiol Biochem 2016; 122:47-53. [PMID: 26707160 DOI: 10.3109/13813455.2015.1122066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The G0/G1 switch gene 2 (G0S2) protein attenuated adipose triglyceride lipase (ATGL) activity and decreased lipolysis in rodent and human adipocytes. We hypothesized that G0S2 mRNA expression in human adipose tissue is influenced by depot, adipocyte size, body weight and caloric intake. Adipose tissue samples were obtained during abdominal surgery and by needle biopsy before and 3 h after an extended glucose load in lean subjects. G0S2 mRNA was 7× higher expressed in mature human adipocytes compared to the stromavascular fraction. Cell size inversely correlated with G0S2 mRNA expression in both, subcutaneous and omental adipose depots. G0S2 mRNA expression was 75% higher in subcutaneous compared to omental adipose tissue. Obesity was associated with lower G0S2 mRNA expression in subcutaneous adipose tissue. Acute glucose ingestion after an overnight fast did not significantly increase G0S2 expression in subcutaneous adipose tissue. In conclusion, differences in G0S2 expression may explain depot-specific and obesity-associated differences in lipolysis on the molecular level.
Collapse
Affiliation(s)
- Alexander Skopp
- a Institute of Clinical Pharmacology, Hannover Medical School , Hannover , Germany
| | - Marcus May
- a Institute of Clinical Pharmacology, Hannover Medical School , Hannover , Germany
| | - Juergen Janke
- b Max-Delbrück Center for Molecular Medicine , Berlin , Germany
| | - Heike Kielstein
- c Department of Anatomy and Cell Biology , Martin Luther University Halle-Wittenberg , Halle (Saale) , Germany , and
| | - Ruth Wunder
- d Surgical Department , Clementinenhaus , Hannover , Germany
| | | | - Andreas Kuthe
- d Surgical Department , Clementinenhaus , Hannover , Germany
| | - Jens Jordan
- a Institute of Clinical Pharmacology, Hannover Medical School , Hannover , Germany
| | - Stefan Engeli
- a Institute of Clinical Pharmacology, Hannover Medical School , Hannover , Germany
| |
Collapse
|
40
|
Heier C, Radner FPW, Moustafa T, Schreiber R, Grond S, Eichmann TO, Schweiger M, Schmidt A, Cerk IK, Oberer M, Theussl HC, Wojciechowski J, Penninger JM, Zimmermann R, Zechner R. G0/G1 Switch Gene 2 Regulates Cardiac Lipolysis. J Biol Chem 2015; 290:26141-50. [PMID: 26350455 PMCID: PMC4646265 DOI: 10.1074/jbc.m115.671842] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Indexed: 12/13/2022] Open
Abstract
The anabolism and catabolism of myocardial triacylglycerol (TAG) stores are important processes for normal cardiac function. TAG synthesis detoxifies and stockpiles fatty acids to prevent lipotoxicity, whereas TAG hydrolysis (lipolysis) remobilizes fatty acids from endogenous storage pools as energy substrates, signaling molecules, or precursors for complex lipids. This study focused on the role of G0/G1 switch 2 (G0S2) protein, which was previously shown to inhibit the principal TAG hydrolase adipose triglyceride lipase (ATGL), in the regulation of cardiac lipolysis. Using wild-type and mutant mice, we show the following: (i) G0S2 is expressed in the heart and regulated by the nutritional status with highest expression levels after re-feeding. (ii) Cardiac-specific overexpression of G0S2 inhibits cardiac lipolysis by direct protein-protein interaction with ATGL. This leads to severe cardiac steatosis. The steatotic hearts caused by G0S2 overexpression are less prone to fibrotic remodeling or cardiac dysfunction than hearts with a lipolytic defect due to ATGL deficiency. (iii) Conversely to the phenotype of transgenic mice, G0S2 deficiency results in a de-repression of cardiac lipolysis and decreased cardiac TAG content. We conclude that G0S2 acts as a potent ATGL inhibitor in the heart modulating cardiac substrate utilization by regulating cardiac lipolysis.
Collapse
Affiliation(s)
- Christoph Heier
- From the Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | - Franz P W Radner
- From the Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | - Tarek Moustafa
- From the Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria, the Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine and
| | - Renate Schreiber
- From the Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | - Susanne Grond
- From the Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | - Thomas O Eichmann
- From the Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | - Martina Schweiger
- From the Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | - Albrecht Schmidt
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria, and
| | - Ines K Cerk
- From the Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | - Monika Oberer
- From the Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | - H-Christian Theussl
- the Transgenic Service, Research Institute of Molecular Pathology (IMP), 1030 Vienna, Austria, and the IMBA Institute of Molecular Biotechnology of the Austrian Academy of Sciences, 1030 Vienna, Austria
| | - Jacek Wojciechowski
- the Transgenic Service, Research Institute of Molecular Pathology (IMP), 1030 Vienna, Austria, and the IMBA Institute of Molecular Biotechnology of the Austrian Academy of Sciences, 1030 Vienna, Austria
| | - Josef M Penninger
- the Transgenic Service, Research Institute of Molecular Pathology (IMP), 1030 Vienna, Austria, and the IMBA Institute of Molecular Biotechnology of the Austrian Academy of Sciences, 1030 Vienna, Austria
| | - Robert Zimmermann
- From the Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | - Rudolf Zechner
- From the Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria,
| |
Collapse
|
41
|
Jaeger D, Schoiswohl G, Hofer P, Schreiber R, Schweiger M, Eichmann TO, Pollak NM, Poecher N, Grabner GF, Zierler KA, Eder S, Kolb D, Radner FPW, Preiss-Landl K, Lass A, Zechner R, Kershaw EE, Haemmerle G. Fasting-induced G0/G1 switch gene 2 and FGF21 expression in the liver are under regulation of adipose tissue derived fatty acids. J Hepatol 2015; 63:437-45. [PMID: 25733154 PMCID: PMC4518503 DOI: 10.1016/j.jhep.2015.02.035] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 02/18/2015] [Accepted: 02/20/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Adipose tissue (AT)-derived fatty acids (FAs) are utilized for hepatic triacylglycerol (TG) generation upon fasting. However, their potential impact as signaling molecules is not established. Herein we examined the role of exogenous AT-derived FAs in the regulation of hepatic gene expression by investigating mice with a defect in AT-derived FA supply to the liver. METHODS Plasma FA levels, tissue TG hydrolytic activities and lipid content were determined in mice lacking the lipase co-activator comparative gene identification-58 (CGI-58) selectively in AT (CGI-58-ATko) applying standard protocols. Hepatic expression of lipases, FA oxidative genes, transcription factors, ER stress markers, hormones and cytokines were determined by qRT-PCR, Western blotting and ELISA. RESULTS Impaired AT-derived FA supply upon fasting of CGI-58-ATko mice causes a marked defect in liver PPARα-signaling and nuclear CREBH translocation. This severely reduced the expression of respective target genes such as the ATGL inhibitor G0/G1 switch gene-2 (G0S2) and the endocrine metabolic regulator FGF21. These changes could be reversed by lipid administration and raising plasma FA levels. Impaired AT-lipolysis failed to induce hepatic G0S2 expression in fasted CGI-58-ATko mice leading to enhanced ATGL-mediated TG-breakdown strongly reducing hepatic TG deposition. On high fat diet, impaired AT-lipolysis counteracts hepatic TG accumulation and liver stress linked to improved systemic insulin sensitivity. CONCLUSIONS AT-derived FAs are a critical regulator of hepatic fasting gene expression required for the induction of G0S2-expression in the liver to control hepatic TG-breakdown. Interfering with AT-lipolysis or hepatic G0S2 expression represents an effective strategy for the treatment of hepatic steatosis.
Collapse
Affiliation(s)
- Doris Jaeger
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Gabriele Schoiswohl
- Division of Endocrinology, Diabetes, and Metabolism, University of Pittsburgh, PA 15261, USA
| | - Peter Hofer
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Renate Schreiber
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Thomas O Eichmann
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Nina M Pollak
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Nadja Poecher
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Gernot F Grabner
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Kathrin A Zierler
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Sandra Eder
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Dagmar Kolb
- ZMF, Center for Medical Research, Medical University of Graz, A-8010 Graz, Austria
| | - Franz P W Radner
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Karina Preiss-Landl
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Erin E Kershaw
- Division of Endocrinology, Diabetes, and Metabolism, University of Pittsburgh, PA 15261, USA
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria.
| |
Collapse
|
42
|
Cidea improves the metabolic profile through expansion of adipose tissue. Nat Commun 2015; 6:7433. [PMID: 26118629 DOI: 10.1038/ncomms8433] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 05/08/2015] [Indexed: 02/06/2023] Open
Abstract
In humans, Cidea (cell death-inducing DNA fragmentation factor alpha-like effector A) is highly but variably expressed in white fat, and expression correlates with metabolic health. Here we generate transgenic mice expressing human Cidea in adipose tissues (aP2-hCidea mice) and show that Cidea is mechanistically associated with a robust increase in adipose tissue expandability. Under humanized conditions (thermoneutrality, mature age and prolonged exposure to high-fat diet), aP2-hCidea mice develop a much more pronounced obesity than their wild-type littermates. Remarkably, the malfunctioning of visceral fat normally caused by massive obesity is fully overcome-perilipin 1 and Akt expression are preserved, tissue degradation is prevented, macrophage accumulation is decreased and adiponectin expression remains high. Importantly, the aP2-hCidea mice display enhanced insulin sensitivity. Our data establish a functional role for Cidea and suggest that, in humans, the association between Cidea levels in white fat and metabolic health is not only correlative but also causative.
Collapse
|
43
|
Lee PH, Yamada T, Park CS, Shen Y, Puppi M, Lacorazza HD. G0S2 modulates homeostatic proliferation of naïve CD8⁺ T cells and inhibits oxidative phosphorylation in mitochondria. Immunol Cell Biol 2015; 93:605-15. [PMID: 25666096 PMCID: PMC4531109 DOI: 10.1038/icb.2015.9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Revised: 12/24/2014] [Accepted: 01/15/2015] [Indexed: 11/16/2022]
Abstract
Since its discovery, diverse functions have been attributed to the G0/G1 switch gene 2 (G0S2), from lipid metabolism to control of cell proliferation. Our group showed for the first time that G0S2 promotes quiescence in hematopoietic stem cells by interacting with and retaining nucleolin around the nucleus. Herein, we report the role of G0S2 in the differentiation and function of CD8+ T cells examined in mice with an embryonic deletion of the G0s2 gene. G0S2 expression in naïve CD8+ T cells decreased immediately after T-cell receptor activation downstream of the MAPK, calcium/calmodulin, PI3K, and mTOR pathways. Surprisingly, G0S2-null naïve CD8+ T cells displayed increased basal and spare respiratory capacity that was not associated with increased mitochondrial biogenesis but with increased phosphorylation of AMPKα. Naïve CD8+ T cells showed increased proliferation in response to in vitro activation and in vivo lymphopenia; however, naïve CD8+ T cells expressing the OT-1 transgene exhibited normal differentiation of naïve cells to effector and memory CD8+ T cells upon infection with Listeria monocytogenes in a wild type or a G0s2-null environment, with increased circulating levels of free fatty acids. Collectively, our results suggest that G0S2 inhibits energy production by oxidative phosphorylation to fine-tune proliferation in homeostatic conditions.
Collapse
Affiliation(s)
- Ping-Hsien Lee
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Takeshi Yamada
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Chun Shik Park
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Ye Shen
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Monica Puppi
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - H Daniel Lacorazza
- 1] Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA [2] Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|
44
|
Wang Y, Zhang Y, Zhu Y, Zhang P. Lipolytic inhibitor G0/G1 switch gene 2 inhibits reactive oxygen species production and apoptosis in endothelial cells. Am J Physiol Cell Physiol 2015; 308:C496-504. [PMID: 25588877 DOI: 10.1152/ajpcell.00317.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
G0/G1 switch gene 2 (G0S2), a novel target gene of peroxisome proliferator-activated receptor, is highly expressed in fat tissues. G0S2 acts as proapoptotic factor toward human cancer cells. Endothelial cell (EC) apoptosis may be an initiating event in the development of atherosclerosis. However, the expression and function of G0S2 in vascular ECs remain unknown. Here, we reported for the first time that G0S2 is expressed in arterial ECs. Ectopic expression of G0S2 increased neutral lipid accumulation in cultured ECs. However, G0S2 prevented ECs from serum-free starvation stress- and hydrogen peroxide (H2O2)-induced apoptosis. G0S2 blocked the H2O2-induced dissipation of mitochondrial membrane potential. G0S2 decreased the release of cytochrome c from mitochondria into the cytosol, followed by activation of caspase-9 and caspase-3. The anti-apoptotic effect of G0S2 was Bcl-2 and adipose triglyceride lipase independent. In contrast, gene silence of G0S2 increased serum-free starvation stress-induced EC apoptosis and decreased the formation of capillary-like structures. We further found that G0S2 couples with the F0F1-ATP synthase in ECs. Levels of ATP were elevated, whereas reactive oxygen species levels were reduced in G0S2-expressing ECs. G0S2 can inhibit endothelial denudation secondary to H2O2-induced injury to ECs in vivo. These results indicate that G0S2 acts as a prosurvival molecule in ECs. Taken together, our results indicate that G0S2 has a protective function in ECs and may be a potential target for the treatment of cardiovascular diseases associated with reactive oxygen species-induced EC injury, such as atherosclerosis and restenosis.
Collapse
Affiliation(s)
- Yinfang Wang
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, China
| | - Yahui Zhang
- Department of Pathophysiology, Hubei University of Medicine, Hubei, China; and
| | - Yichun Zhu
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, China
| | - Peng Zhang
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
45
|
Heier C, Zimmermann R. The sparing use of fat: G0s2 controls lipolysis and fatty acid oxidation. Diabetologia 2015; 58:7-9. [PMID: 25351607 DOI: 10.1007/s00125-014-3430-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 10/06/2014] [Indexed: 10/24/2022]
Affiliation(s)
- Christoph Heier
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31A, 8010, Graz, Austria
| | | |
Collapse
|
46
|
El-Assaad W, El-Kouhen K, Mohammad AH, Yang J, Morita M, Gamache I, Mamer O, Avizonis D, Hermance N, Kersten S, Tremblay ML, Kelliher MA, Teodoro JG. Deletion of the gene encoding G0/G 1 switch protein 2 (G0s2) alleviates high-fat-diet-induced weight gain and insulin resistance, and promotes browning of white adipose tissue in mice. Diabetologia 2015; 58:149-57. [PMID: 25381555 PMCID: PMC5001162 DOI: 10.1007/s00125-014-3429-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 09/29/2014] [Indexed: 11/29/2022]
Abstract
AIMS/HYPOTHESIS Obesity is a global epidemic resulting from increased energy intake, which alters energy homeostasis and results in an imbalance in fat storage and breakdown. G0/G1 switch gene 2 (G0s2) has been recently characterised in vitro as an inhibitor of adipose triglyceride lipase (ATGL), the rate-limiting step in fat catabolism. In the current study we aim to functionally characterise G0s2 within the physiological context of a mouse model. METHODS We generated a mouse model in which G0s2 was deleted. The homozygous G0s2 knockout (G0s2 (-/-)) mice were studied over a period of 22 weeks. Metabolic variables were measured including body weight and body composition, food intake, glucose and insulin tolerance tests, energy metabolism and thermogenesis. RESULTS We report that G0s2 inhibits ATGL and regulates lipolysis and energy metabolism in vivo. G0s2 (-/-) mice are lean, resistant to weight gain induced by a high-fat diet and are glucose tolerant and insulin sensitive. The white adipose tissue of G0s2 (-/-) mice has enhanced lipase activity and adipocytes showed enhanced stimulated lipolysis. Energy metabolism in the G0s2 (-/-) mice is shifted towards enhanced lipid metabolism and increased thermogenesis. G0s2 (-/-) mice showed enhanced cold tolerance and increased expression of thermoregulatory and oxidation genes within white adipose tissue, suggesting enhanced 'browning' of the white adipose tissue. CONCLUSIONS/INTERPRETATION Our data show that G0s2 is a physiological regulator of adiposity and energy metabolism and is a potential target in the treatment of obesity and insulin resistance.
Collapse
Affiliation(s)
- Wissal El-Assaad
- Goodman Cancer Research Centre, McGill University, Montréal, QC, Canada
| | - Karim El-Kouhen
- Goodman Cancer Research Centre, McGill University, Montréal, QC, Canada
| | - Amro H. Mohammad
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| | - Jieyi Yang
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| | - Masahiro Morita
- Goodman Cancer Research Centre, McGill University, Montréal, QC, Canada
| | - Isabelle Gamache
- Goodman Cancer Research Centre, McGill University, Montréal, QC, Canada
| | - Orval Mamer
- Metabolomics Core Facility, Goodman Cancer Research Centre, McGill University, Montréal, QC, Canada
| | - Daina Avizonis
- Metabolomics Core Facility, Goodman Cancer Research Centre, McGill University, Montréal, QC, Canada
| | - Nicole Hermance
- Department of Cancer Biology, University of Massachusetts, Worcester, MA, USA
| | - Sander Kersten
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, the Netherlands
| | - Michel L. Tremblay
- Goodman Cancer Research Centre, McGill University, Montréal, QC, Canada
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| | | | - Jose G. Teodoro
- Goodman Cancer Research Centre, McGill University, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| |
Collapse
|
47
|
Cerk IK, Salzburger B, Boeszoermenyi A, Heier C, Pillip C, Romauch M, Schweiger M, Cornaciu I, Lass A, Zimmermann R, Zechner R, Oberer M. A peptide derived from G0/G1 switch gene 2 acts as noncompetitive inhibitor of adipose triglyceride lipase. J Biol Chem 2014; 289:32559-70. [PMID: 25258314 PMCID: PMC4239610 DOI: 10.1074/jbc.m114.602599] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The protein G0/G1 switch gene 2 (G0S2) is a small basic protein that functions as an endogenous inhibitor of adipose triglyceride lipase (ATGL), a key enzyme in intracellular lipolysis. In this study, we identified a short sequence covering residues Lys-20 to Ala-52 in G0S2 that is still fully capable of inhibiting mouse and human ATGL. We found that a synthetic peptide corresponding to this region inhibits ATGL in a noncompetitive manner in the nanomolar range. This peptide is highly selective for ATGL and does not inhibit other lipases, including hormone-sensitive lipase, monoacylglycerol lipase, lipoprotein lipase, and patatin domain-containing phospholipases 6 and 7. Because increased lipolysis is linked to the development of metabolic disorders, the inhibition of ATGL by G0S2-derived peptides may represent a novel therapeutic tool to modulate lipolysis.
Collapse
Affiliation(s)
- Ines K Cerk
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Barbara Salzburger
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Andras Boeszoermenyi
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Christoph Heier
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Christoph Pillip
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Matthias Romauch
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Martina Schweiger
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Irina Cornaciu
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Achim Lass
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Robert Zimmermann
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Rudolf Zechner
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Monika Oberer
- From the Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| |
Collapse
|
48
|
McGowan SE, McCoy DM. Regulation of fibroblast lipid storage and myofibroblast phenotypes during alveolar septation in mice. Am J Physiol Lung Cell Mol Physiol 2014; 307:L618-31. [PMID: 25150063 DOI: 10.1152/ajplung.00144.2014] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Signaling through platelet-derived growth factor receptor-α (PDGFRα) is required for alveolar septation and participates in alveolar regeneration after pneumonectomy. In both adipose tissue and skeletal muscle, bipotent pdgfrα-expressing progenitors expressing delta-like ligand-1 or sex-determining region Y box 9 (Sox9) may differentiate into either lipid storage cells or myofibroblasts. We analyzed markers of mesenchymal progenitors and differentiation in lung fibroblasts (LF) with different levels (absent, low, or high) of pdgfrα gene expression. A larger proportion of pdgfrα-expressing than nonexpressing LF contained Sox9. Neutral lipids, CD166, and Tcf21 were more abundant in LF with a lower compared with a higher level of pdgfrα gene expression. PDGF-A increased Sox9 in primary LF cultures, suggesting that active signaling through PDGFRα is required to maintain Sox9. As alveolar septation progresses from postnatal day (P) 8 to P12, fewer pdgfrα-expressing LF contain Sox9, whereas more of these LF contain myocardin-like transcription factor-A, showing that Sox9 diminishes as LF become myofibroblasts. At P8, neutral lipid droplets predominate in LF with the lower level of pdgfrα gene expression, whereas transgelin (tagln) was predominantly expressed in LF with higher pdgfrα gene expression. Targeted deletion of pdgfrα in LF, which expressed tagln, reduced Sox9 in α-actin (α-SMA, ACTA2)-containing LF, whereas it increased the abundance of cell surface delta-like protein-1 (as well as peroxisome proliferator-activated receptor-γ and tcf21 mRNA in LF, which also expressed stem cell antigen-1). Thus pdgfrα deletion differentially alters delta-like protein-1 and Sox9, suggesting that targeting different downstream pathways in PDGF-A-responsive LF could identify strategies that promote lung regeneration without initiating fibrosis.
Collapse
Affiliation(s)
- Stephen E McGowan
- Department of Veterans Affairs Research Service and Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Diann M McCoy
- Department of Veterans Affairs Research Service and Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
49
|
Nielsen TS, Jessen N, Jørgensen JOL, Møller N, Lund S. Dissecting adipose tissue lipolysis: molecular regulation and implications for metabolic disease. J Mol Endocrinol 2014; 52:R199-222. [PMID: 24577718 DOI: 10.1530/jme-13-0277] [Citation(s) in RCA: 278] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lipolysis is the process by which triglycerides (TGs) are hydrolyzed to free fatty acids (FFAs) and glycerol. In adipocytes, this is achieved by sequential action of adipose TG lipase (ATGL), hormone-sensitive lipase (HSL), and monoglyceride lipase. The activity in the lipolytic pathway is tightly regulated by hormonal and nutritional factors. Under conditions of negative energy balance such as fasting and exercise, stimulation of lipolysis results in a profound increase in FFA release from adipose tissue (AT). This response is crucial in order to provide the organism with a sufficient supply of substrate for oxidative metabolism. However, failure to efficiently suppress lipolysis when FFA demands are low can have serious metabolic consequences and is believed to be a key mechanism in the development of type 2 diabetes in obesity. As the discovery of ATGL in 2004, substantial progress has been made in the delineation of the remarkable complexity of the regulatory network controlling adipocyte lipolysis. Notably, regulatory mechanisms have been identified on multiple levels of the lipolytic pathway, including gene transcription and translation, post-translational modifications, intracellular localization, protein-protein interactions, and protein stability/degradation. Here, we provide an overview of the recent advances in the field of AT lipolysis with particular focus on the molecular regulation of the two main lipases, ATGL and HSL, and the intracellular and extracellular signals affecting their activity.
Collapse
Affiliation(s)
- Thomas Svava Nielsen
- The Novo Nordisk Foundation Center for Basic Metabolic ResearchSection on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 6.6.30, DK-2200 N Copenhagen, DenmarkDepartment of Endocrinology and Internal MedicineAarhus University Hospital, Nørrebrogade 44, Bldg. 3.0, 8000 Aarhus C, DenmarkDepartment of Molecular MedicineAarhus University Hospital, Brendstrupgårdsvej 100, 8200 Aarhus N, DenmarkThe Novo Nordisk Foundation Center for Basic Metabolic ResearchSection on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 6.6.30, DK-2200 N Copenhagen, DenmarkDepartment of Endocrinology and Internal MedicineAarhus University Hospital, Nørrebrogade 44, Bldg. 3.0, 8000 Aarhus C, DenmarkDepartment of Molecular MedicineAarhus University Hospital, Brendstrupgårdsvej 100, 8200 Aarhus N, Denmark
| | - Niels Jessen
- The Novo Nordisk Foundation Center for Basic Metabolic ResearchSection on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 6.6.30, DK-2200 N Copenhagen, DenmarkDepartment of Endocrinology and Internal MedicineAarhus University Hospital, Nørrebrogade 44, Bldg. 3.0, 8000 Aarhus C, DenmarkDepartment of Molecular MedicineAarhus University Hospital, Brendstrupgårdsvej 100, 8200 Aarhus N, DenmarkThe Novo Nordisk Foundation Center for Basic Metabolic ResearchSection on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 6.6.30, DK-2200 N Copenhagen, DenmarkDepartment of Endocrinology and Internal MedicineAarhus University Hospital, Nørrebrogade 44, Bldg. 3.0, 8000 Aarhus C, DenmarkDepartment of Molecular MedicineAarhus University Hospital, Brendstrupgårdsvej 100, 8200 Aarhus N, Denmark
| | - Jens Otto L Jørgensen
- The Novo Nordisk Foundation Center for Basic Metabolic ResearchSection on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 6.6.30, DK-2200 N Copenhagen, DenmarkDepartment of Endocrinology and Internal MedicineAarhus University Hospital, Nørrebrogade 44, Bldg. 3.0, 8000 Aarhus C, DenmarkDepartment of Molecular MedicineAarhus University Hospital, Brendstrupgårdsvej 100, 8200 Aarhus N, Denmark
| | - Niels Møller
- The Novo Nordisk Foundation Center for Basic Metabolic ResearchSection on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 6.6.30, DK-2200 N Copenhagen, DenmarkDepartment of Endocrinology and Internal MedicineAarhus University Hospital, Nørrebrogade 44, Bldg. 3.0, 8000 Aarhus C, DenmarkDepartment of Molecular MedicineAarhus University Hospital, Brendstrupgårdsvej 100, 8200 Aarhus N, Denmark
| | - Sten Lund
- The Novo Nordisk Foundation Center for Basic Metabolic ResearchSection on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 6.6.30, DK-2200 N Copenhagen, DenmarkDepartment of Endocrinology and Internal MedicineAarhus University Hospital, Nørrebrogade 44, Bldg. 3.0, 8000 Aarhus C, DenmarkDepartment of Molecular MedicineAarhus University Hospital, Brendstrupgårdsvej 100, 8200 Aarhus N, Denmark
| |
Collapse
|