151
|
Zhao NQ, Li XY, Wang L, Feng ZL, Li XF, Wen YF, Han JX. Palmitate induces fat accumulation by activating C/EBPβ-mediated G0S2 expression in HepG2 cells. World J Gastroenterol 2017; 23:7705-7715. [PMID: 29209111 PMCID: PMC5703930 DOI: 10.3748/wjg.v23.i43.7705] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 09/27/2017] [Accepted: 09/28/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To determine the role of G0/G1 switch gene 2 (G0S2) and its transcriptional regulation in palmitate-induced hepatic lipid accumulation.
METHODS HepG2 cells were treated with palmitate, or palmitate in combination with CCAAT/enhancer binding protein (C/EBP)β siRNA or G0S2 siRNA. The mRNA expression of C/EBPβ, peroxisome proliferator-activated receptor (PPAR)γ and PPARγ target genes (G0S2, GPR81, GPR109A and Adipoq) was examined by qPCR. The protein expression of C/EBPβ, PPARγ, and G0S2 was determined by Western blotting. Lipid accumulation was detected with Oil Red O staining and quantified by absorbance value of the extracted Oil Red O dye. Lipolysis was evaluated by measuring the amount of glycerol released into the medium.
RESULTS Palmitate caused a dose-dependent increase in lipid accumulation and a dose-dependent decrease in lipolysis in HepG2 cells. In addition, palmitate increased the mRNA expression of C/EBPβ, PPARγ, and PPARγ target genes (G0S2, GPR81, GPR109A, and Adipoq) and the protein expression of C/EBPβ, PPARγ, and G0S2 in a dose-dependent manner. Knockdown of C/EBPβ decreased palmitate-induced PPARγ and its target genes (G0S2, GPR81, GPR109A, and Adipoq) mRNA expression and palmitate-induced PPARγ and G0S2 protein expression in HepG2 cells. Knockdown of C/EBPβ also attenuated lipid accumulation and augmented lipolysis in palmitate-treated HepG2 cells. G0S2 knockdown attenuated lipid accumulation and augmented lipolysis, while G0S2 knockdown had no effects on the mRNA expression of C/EBPβ, PPARγ, and PPARγ target genes (GPR81, GPR109A and Adipoq) in palmitate-treated HepG2 cells.
CONCLUSION Palmitate can induce lipid accumulation in HepG2 cells by activating C/EBPβ-mediated G0S2 expression.
Collapse
Affiliation(s)
- Nai-Qian Zhao
- Department of Gerontology, the Second Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Xiao-Yan Li
- Department of Infectious Diseases, the First People’s Hospital of Jinzhong, Jinzhong 030600, Shanxi Province, China
| | - Li Wang
- Department of Gerontology, the Second Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Zi-Ling Feng
- Department of Infectious Diseases, the First People’s Hospital of Jinzhong, Jinzhong 030600, Shanxi Province, China
| | - Xi-Fen Li
- Department of Infectious Diseases, the First People’s Hospital of Jinzhong, Jinzhong 030600, Shanxi Province, China
| | - Yan-Fang Wen
- Department of Infectious Diseases, the First People’s Hospital of Jinzhong, Jinzhong 030600, Shanxi Province, China
| | - Jin-Xiang Han
- Department of Gerontology, the Second Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| |
Collapse
|
152
|
Qin S, Han H, Zhang K, Ding X, Bai S, Wang J, Zeng Q. Dietary fibre alleviates hepatic fat deposition via inhibiting lipogenic gene expression in meat ducks. J Anim Physiol Anim Nutr (Berl) 2017; 102:e736-e745. [DOI: 10.1111/jpn.12828] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/01/2017] [Indexed: 02/03/2023]
Affiliation(s)
- S. Qin
- Institute of Animal Nutrition; Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education; Sichuan Agricultural University; Chengdu China
| | - H. Han
- Institute of Animal Nutrition; Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education; Sichuan Agricultural University; Chengdu China
| | - K. Zhang
- Institute of Animal Nutrition; Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education; Sichuan Agricultural University; Chengdu China
| | - X. Ding
- Institute of Animal Nutrition; Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education; Sichuan Agricultural University; Chengdu China
| | - S. Bai
- Institute of Animal Nutrition; Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education; Sichuan Agricultural University; Chengdu China
| | - J. Wang
- Institute of Animal Nutrition; Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education; Sichuan Agricultural University; Chengdu China
| | - Q. Zeng
- Institute of Animal Nutrition; Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education; Sichuan Agricultural University; Chengdu China
| |
Collapse
|
153
|
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: 377] [Impact Index Per Article: 47.1] [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
|
154
|
Liu B, Jiang H, Lu J, Baiyun R, Li S, Lv Y, Li D, Wu H, Zhang Z. Grape seed procyanidin extract ameliorates lead-induced liver injury via miRNA153 and AKT/GSK-3β/Fyn-mediated Nrf2 activation. J Nutr Biochem 2017; 52:115-123. [PMID: 29175668 DOI: 10.1016/j.jnutbio.2017.09.025] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/02/2017] [Accepted: 09/28/2017] [Indexed: 12/12/2022]
Abstract
Lead-induced hepatotoxicity is characterized by an extensive oxidative stress. Grape seed procyanidin extract (GSPE) possesses abundant biological activities. Herein, we investigated the protective role of GSPE against lead-induced liver injury and determined the potential molecular mechanisms. In vivo, rats were treated with/without lead acetate (PbAc) (0.05%, w/v) in the presence/absence of GSPE (200 mg/kg). In vitro, hepatocytes were pretreated with/without GSPE (100 μg/ml) in the presence/absence of PbAc (100 μM). PbAc administration to rats resulted in anemia, liver dysfunction, lead accumulation in the bone and liver, oxidative stress, DNA damage and apoptosis. GSPE significantly attenuated these adverse effects, except lead accumulation in liver. GSPE also decreased the expression of miRNA153 and increased the translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and levels of its downstream protein, and protein kinase B (AKT) phosphorylation in PbAc-induced liver injury. In primary hepatocytes treated with PbAc, GSPE increased hepatocyte viability and decreased lactate dehydrogenase release and reactive oxygen species levels. Dietary GSPE attenuated PbAc-induced liver injury in rats via an integrated mechanism associated with the miRNA153 and AKT/glycogen synthase kinase 3 beta/Fyn-mediated Nrf2 activation.
Collapse
Affiliation(s)
- Biying Liu
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China
| | - Huijie Jiang
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China
| | - Jingjing Lu
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China
| | - Ruiqi Baiyun
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China
| | - Yueying Lv
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China
| | - Da Li
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China
| | - Hao Wu
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China.
| |
Collapse
|
155
|
Lee SM, Dorotea D, Jung I, Nakabayashi T, Miyata T, Ha H. TM5441, a plasminogen activator inhibitor-1 inhibitor, protects against high fat diet-induced non-alcoholic fatty liver disease. Oncotarget 2017; 8:89746-89760. [PMID: 29163785 PMCID: PMC5685706 DOI: 10.18632/oncotarget.21120] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 09/03/2017] [Indexed: 12/16/2022] Open
Abstract
Recent evidences showed that elevation of plasminogen activator inhibitor 1 (PAI-1) was responsible in mediating obesity-induced non-alcoholic fatty liver disease (NAFLD) and metabolic disorders. Here, we investigated the effect of TM5441, an oral PAI-1 inhibitor that lacks of bleeding risk, on high-fat diet (HFD)-induced NAFLD. HFD-fed C57BL/6J mice was daily treated with 20 mg/kg TM5441. To examine the preventive effect, 10-week-treatment was started along with initiation of HFD; alternatively, 4-week-treatment was started in mice with glucose intolerance in the interventional strategy. In vivo study showed that early and delayed treatment decreased hepatic steatosis. Particularly, early treatment prevented the progression of hepatic inflammation and fibrosis in HFD mice. Interestingly, both strategies abrogated hepatic insulin resistance and mitochondrial dysfunction, presented by enhanced p-Akt and p-GSK3β, reduced p-JNK signaling, along with p-AMPK and PGC-1α activation. Consistently, TM5441 treatment in the presence of either PAI-1 exposure or TNF-α stimulated-PAI-1 activity showed a restoration of mitochondrial biogenesis related genes expression on HepG2 cells. Thus, improvement of insulin sensitivity and mitochondrial function was imperative to partially explain the therapeutic effects of TM5441, a novel agent targeting HFD-induced NAFLD.
Collapse
Affiliation(s)
- Seon Myeong Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Debra Dorotea
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Inji Jung
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Tetsuo Nakabayashi
- United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Toshio Miyata
- United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
156
|
Wallstab C, Eleftheriadou D, Schulz T, Damm G, Seehofer D, Borlak J, Holzhütter HG, Berndt N. A unifying mathematical model of lipid droplet metabolism reveals key molecular players in the development of hepatic steatosis. FEBS J 2017; 284:3245-3261. [PMID: 28763157 DOI: 10.1111/febs.14189] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 06/02/2017] [Accepted: 07/28/2017] [Indexed: 12/16/2022]
Abstract
The liver responds to elevated plasma concentrations of free fatty acids (FFAs) with an enhanced uptake of FFAs and their esterification to triacylglycerol (TAG). On the long term, this may result in massive hepatic TAG accumulation called steatosis hepatitis. In hepatocytes, the poor water-soluble TAG is packed in specialized organelles: Lipid droplets (LDs) serving as transient cellular deposit and lipoproteins (LPs) transporting TAG and cholesterol esters to extra-hepatic tissues. The dynamics of these organelles is controlled by a variety of regulatory surface proteins (RSPs). Assembly and export of VLDLs are mainly regulated by the microsomal transfer protein (MTP) and apoprotein B100. Formation and lipolysis of LDs are regulated by several RSPs. The best studied regulators belong to the PAT (Perilipin/Adipophilin/TIP47) and CIDE families. Knockdown or overexpression of SRPs may significantly affect the total number and size distribution of LDs. Intriguingly, a large cell-to-cell heterogeneity with respect to the number and size of LDs has been found in various cell types including hepatocytes. These findings suggest that the extent of cellular lipid accumulation is determined not only by the imbalance between lipid supply and utilization but also by variations in the expression of RSPs and metabolic enzymes. To better understand the relative regulatory impact of individual processes involved in the cellular TAG turnover, we developed a comprehensive kinetic model encompassing the pathways of the fatty acid and triglyceride metabolism and the main molecular processes governing the dynamics of LDs. The model was parametrized such that a large number of experimental in vitro and in vivo findings are correctly recapitulated. A control analysis of the model revealed that variations in the activity of FFA uptake, diacylglycerol acyltransferase (DGAT) 2, and adipose triglyceride lipase (ATGL) have the strongest influence on the cellular TAG level. We used the model to simulate LD size distributions in human hepatoma cells and hepatocytes exposed to a challenge with FFAs. A random fold change by a factor of about two in the activity of RSPs was sufficient to reproduce the large diversity of droplet size distributions observed in individual cells. Under the premise that the same extent of variability of RSPs holds for the intact organ, our model predicts variations in the TAG content of individual hepatocytes by a factor of about 3-6 depending on the nutritional regime. Taken together, our modeling approach integrates numerous experimental findings on individual processes in the cellular TAG metabolism and LD dynamics metabolism to a consistent state-of-the-art dynamic network model that can be used to study how changes in the external conditions or systemic parameters will affect the TAG content of hepatocytes.
Collapse
Affiliation(s)
- Christin Wallstab
- Institute of Biochemistry, Computational Systems Biochemistry Group, Charite - University Medicine Berlin, Germany
| | - Dimitra Eleftheriadou
- Institute of Biochemistry, Computational Systems Biochemistry Group, Charite - University Medicine Berlin, Germany
| | - Theresa Schulz
- Clinic for General-, Visceral- and Transplantation Surgery, Charite - University Medicine Berlin, Germany
| | - Georg Damm
- Clinic for General-, Visceral- and Transplantation Surgery, Charite - University Medicine Berlin, Germany.,Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, Germany
| | - Daniel Seehofer
- Clinic for General-, Visceral- and Transplantation Surgery, Charite - University Medicine Berlin, Germany.,Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, Germany
| | - Jürgen Borlak
- Centre for Pharmacology and Toxicology, Institute for Pharmaco- and Toxicogenomics, Hannover Medical School, Hannover, Germany
| | - Hermann-Georg Holzhütter
- Institute of Biochemistry, Computational Systems Biochemistry Group, Charite - University Medicine Berlin, Germany
| | - Nikolaus Berndt
- Institute of Biochemistry, Computational Systems Biochemistry Group, Charite - University Medicine Berlin, Germany
| |
Collapse
|
157
|
Ploeger JM, Manivel JC, Boatner LN, Mashek DG. Caloric Restriction Prevents Carcinogen-Initiated Liver Tumorigenesis in Mice. Cancer Prev Res (Phila) 2017; 10:660-670. [DOI: 10.1158/1940-6207.capr-17-0174] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 07/27/2017] [Accepted: 08/22/2017] [Indexed: 11/16/2022]
|
158
|
Maternal Prenatal Folic Acid Supplementation Programs Offspring Lipid Metabolism by Aberrant DNA Methylation in Hepatic ATGL and Adipose LPL in Rats. Nutrients 2017; 9:nu9090935. [PMID: 28846595 PMCID: PMC5622695 DOI: 10.3390/nu9090935] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/09/2017] [Accepted: 08/23/2017] [Indexed: 12/27/2022] Open
Abstract
The effects of maternal prenatal folic acid supplementation (FAS) on offspring lipid metabolism in adulthood remains unclear, although prenatal FAS is compulsively suggested in many countries. Female Sprague-Dawley rats were fed with control (CON) or FAS diets before and during pregnancy. Male offspring of CON and FAS dams were further divided into two groups at seven weeks for CON and high-fat (HF) diet interventions for eight weeks in adulthood (n = 10). The interactive effects of maternal prenatal FAS and offspring HF in adulthood on lipid metabolism and DNA methylation of genes involved in lipids metabolism were assessed. The male offspring of FAS dams had elevated serum and liver triglyceride level when fed with HF compared to the male offspring of CON dams. The mRNA and protein expression levels of hepatic ATGL and adipose LPL were significantly decreased in offspring of FAS dams than in offspring of CON dams. Furthermore, maternal prenatal FAS resulted in elevated DNA methylation levels in the promoter and first exon region of hepatic ATGL and adipose LPL in offspring. Maternal FAS exacerbated the adverse effects of HF on lipid metabolism in offspring through inducing aberrant DNA methylation levels of hepatic ATGL and adipose LPL.
Collapse
|
159
|
Franklin MP, Sathyanarayan A, Mashek DG. Acyl-CoA Thioesterase 1 (ACOT1) Regulates PPARα to Couple Fatty Acid Flux With Oxidative Capacity During Fasting. Diabetes 2017; 66:2112-2123. [PMID: 28607105 PMCID: PMC5521868 DOI: 10.2337/db16-1519] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 05/17/2017] [Indexed: 12/18/2022]
Abstract
Hepatic acyl-CoA thioesterase 1 (ACOT1) catalyzes the conversion of acyl-CoAs to fatty acids (FAs) and CoA. We sought to determine the role of ACOT1 in hepatic lipid metabolism in C57Bl/6J male mice 1 week after adenovirus-mediated Acot1 knockdown. Acot1 knockdown reduced liver triglyceride (TG) as a result of enhanced TG hydrolysis and subsequent FA oxidation. In vitro experiments demonstrated that Acot1 knockdown led to greater TG turnover and FA oxidation, suggesting that ACOT1 is important for controlling the rate of FA oxidation. Despite increased FA oxidation, Acot1 knockdown reduced the expression of peroxisome proliferator-activated receptor α (PPARα) target genes, whereas overexpression increased PPARα reporter activity, suggesting ACOT1 regulates PPARα by producing FA ligands. Moreover, ACOT1 exhibited partial nuclear localization during fasting and cAMP/cAMP-dependent protein kinase signaling, suggesting local regulation of PPARα. As a consequence of increased FA oxidation and reduced PPARα activity, Acot1 knockdown enhanced hepatic oxidative stress and inflammation. The effects of Acot1 knockdown on PPARα activity, oxidative stress, and inflammation were rescued by supplementation with Wy-14643, a synthetic PPARα ligand. We demonstrate through these results that ACOT1 regulates fasting hepatic FA metabolism by balancing oxidative flux and capacity.
Collapse
Affiliation(s)
- Mallory P Franklin
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| | | | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN
| |
Collapse
|
160
|
Sugaya Y, Satoh H. Liver-specific G 0 /G 1 switch gene 2 (G0s2) expression promotes hepatic insulin resistance by exacerbating hepatic steatosis in male Wistar rats. J Diabetes 2017; 9:754-763. [PMID: 27624922 DOI: 10.1111/1753-0407.12482] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/18/2016] [Accepted: 09/08/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Hepatic steatosis is strongly associated with insulin resistance. It has been reported that G0 /G1 switch gene 2 (G0s2) inhibits the lipolytic activity of adipose triglyceride lipase, which is a major lipase in the liver as well as in adipocytes. Moreover, G0s2 protein content is increased in the livers of high-fat diet (HFD)-fed rats. In the present study, we investigated the effect of hepatic G0s2 on insulin sensitivity in male Wistar rats. METHODS Male Wistar rats were fed a 60% HFD for 4 weeks. After 3 weeks of feeding, rats were injected with adenovirus-expressing green fluorescent protein (Ad-GFP; control) or adenovirus-expressing mouse G0s2 (Ad-G0s2). On Day 7 after injection, a euglycemic-hyperinsulinemic clamp study was performed in rats fasted for 8 h. RESULTS Body weight and fasting glucose levels were not significantly different between the Ad-GFP and Ad-G0s2 groups. During the clamp study, the glucose infusion rate required for euglycemia decreased significantly by 16% in the Ad-G0s2 compared with Ad-GFP group. The insulin-suppressed hepatic glucose output increased significantly in the Ad-G0s2 group, but the insulin-stimulated glucose disposal rate was not significantly different between the two groups. Consistent with the clamp data, insulin-stimulated phosphorylation of Akt decreased significantly in livers of rats injected with Ad-G0s2. Furthermore, Oil Red O-staining indicated that overexpression of G0s2 protein in the liver promoted hepatic steatosis by 2.5-fold in HFD-fed rats. CONCLUSION The results of the present study indicate that hepatic G0s2 protein may promote hepatic insulin resistance by exacerbating hepatic steatosis.
Collapse
Affiliation(s)
- Yoshiyuki Sugaya
- Department of Nephrology, Hypertension, Diabetology, Endocrinology, and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Hiroaki Satoh
- Department of Nephrology, Hypertension, Diabetology, Endocrinology, and Metabolism, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
161
|
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
|
162
|
Schulze RJ, Sathyanarayan A, Mashek DG. Breaking fat: The regulation and mechanisms of lipophagy. Biochim Biophys Acta Mol Cell Biol Lipids 2017. [PMID: 28642194 DOI: 10.1016/j.bbalip.2017.06.008] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lipophagy is defined as the autophagic degradation of intracellular lipid droplets (LDs). While the field of lipophagy research is relatively young, an expansion of research in this area over the past several years has greatly advanced our understanding of lipophagy. Since its original characterization in fasted liver, the contribution of lipophagy is now recognized in various organisms, cell types, metabolic states and disease models. Moreover, recent studies provide exciting new insights into the underlying mechanisms of lipophagy induction as well as the consequences of lipophagy on cell metabolism and signaling. This review summarizes recent work focusing on LDs and lipophagy as well as highlighting challenges and future directions of research as our understanding of lipophagy continues to grow and evolve. 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)
- Ryan J Schulze
- Department of Biochemistry and Molecular Biology and the Center for Digestive Diseases, Mayo Clinic, Rochester, MN, United States
| | - Aishwarya Sathyanarayan
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States; Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
163
|
Schulze RJ, Drižytė K, Casey CA, McNiven MA. Hepatic Lipophagy: New Insights into Autophagic Catabolism of Lipid Droplets in the Liver. Hepatol Commun 2017; 1:359-369. [PMID: 29109982 PMCID: PMC5669271 DOI: 10.1002/hep4.1056] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The liver is a central fat‐storage organ, making it especially susceptible to steatosis as well as subsequent inflammation and cirrhosis. The mechanisms by which the liver mobilizes stored lipid for energy production, however, remain incompletely defined. The catabolic process of autophagy, a well‐known process of bulk cytoplasmic recycling and cellular self‐regeneration, is a central regulator of lipid metabolism in the liver. In the past decade, numerous studies have examined a selective form of autophagy that specifically targets a unique neutral lipid storage organelle, the lipid droplet, to better understand the function for this process in hepatocellular fatty acid metabolism. In the liver (and other oxidative tissues), this specialized pathway, lipophagy, likely plays as important a role in lipid turnover as conventional lipase‐driven lipolysis. In this review, we highlight several recent studies that have contributed to our understanding about the regulation and effects of hepatic lipophagy. (Hepatology Communications 2017;1:359–369)
Collapse
Affiliation(s)
- Ryan J Schulze
- Department of Biochemistry and Molecular Biology and the Center for Digestive Diseases, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Kristina Drižytė
- Department of Biochemistry and Molecular Biology and the Center for Digestive Diseases, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Biochemistry and Molecular Biology Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| | - Carol A Casey
- Department of Internal Medicine, University of Nebraska Medical Center, 988090 Nebraska Medical Center, Omaha, NE, 68198, USA.,Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), Omaha, NE, 68198, USA
| | - Mark A McNiven
- Department of Biochemistry and Molecular Biology and the Center for Digestive Diseases, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| |
Collapse
|
164
|
Gluchowski NL, Becuwe M, Walther TC, Farese RV. Lipid droplets and liver disease: from basic biology to clinical implications. Nat Rev Gastroenterol Hepatol 2017; 14:343-355. [PMID: 28428634 PMCID: PMC6319657 DOI: 10.1038/nrgastro.2017.32] [Citation(s) in RCA: 450] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lipid droplets are dynamic organelles that store neutral lipids during times of energy excess and serve as an energy reservoir during deprivation. Many prevalent metabolic diseases, such as the metabolic syndrome or obesity, often result in abnormal lipid accumulation in lipid droplets in the liver, also called hepatic steatosis. Obesity-related steatosis, or NAFLD in particular, is a major public health concern worldwide and is frequently associated with insulin resistance and type 2 diabetes mellitus. Here, we review the latest insights into the biology of lipid droplets and their role in maintaining lipid homeostasis in the liver. We also offer a perspective of liver diseases that feature lipid accumulation in these lipid storage organelles, which include NAFLD and viral hepatitis. Although clinical applications of this knowledge are just beginning, we highlight new opportunities for identifying molecular targets for treating hepatic steatosis and steatohepatitis.
Collapse
Affiliation(s)
- Nina L. Gluchowski
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, 655 Huntington Avenue, Boston, Massachusetts 02115, USA.,Boston Children’s Hospital Department of Gastroenterology, Hepatology and Nutrition, 300 Longwood Avenue Boston, Massachusetts 02115, USA
| | - Michel Becuwe
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, 655 Huntington Avenue, Boston, Massachusetts 02115, USA
| | - Tobias C. Walther
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, 655 Huntington Avenue, Boston, Massachusetts 02115, USA.,Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue Boston, Massachusetts 02115, USA.,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur Boston, Massachusetts 02115, USA.,Howard Hughes Medical Institute, Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, 655 Huntington Avenue, Boston, Massachusetts 02115, USA
| | - Robert V. Farese
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, 655 Huntington Avenue, Boston, Massachusetts 02115, USA.,Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue Boston, Massachusetts 02115, USA.,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur Boston, Massachusetts 02115, USA
| |
Collapse
|
165
|
Iannucci LF, Cioffi F, Senese R, Goglia F, Lanni A, Yen PM, Sinha RA. Metabolomic analysis shows differential hepatic effects of T 2 and T 3 in rats after short-term feeding with high fat diet. Sci Rep 2017; 7:2023. [PMID: 28515456 PMCID: PMC5435676 DOI: 10.1038/s41598-017-02205-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/07/2017] [Indexed: 01/16/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major health problem worldwide, and is often associated with lipotoxic injury, defective mitochondrial function, and insulin resistance. Thyroid hormones (THs) are important regulators of hepatic lipid metabolism. Among the THs, diiodothyronine (T2) and triiodothyronine (T3) have shown promising results in lowering hepatic fat content in various models of NAFLD. In this study, we used a targeted metabolomics approach to investigate the differential effects of T2 and T3 on the early metabolic adaptation in the livers of rats fed high fat diet (HFD), a period when hepatosteatosis is reversible. Our results showed that both T2 and T3 strongly induced autophagy and intra-hepatic acylcarnitine flux but prevented the generation of sphingolipid/ceramides in animals fed HFD. Interestingly, although both T2 and T3 decreased hepatic fat content, only T2 was able to rescue the impairment in AKT and MAPK/ERK pathways caused by HFD. In summary, we have identified and characterized the effects of T2 and T3 on hepatic metabolism during short-term exposure to HFD. These findings illuminate the common and divergent metabolic pathways by T2 and T3 that also may be important in the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Liliana F Iannucci
- Program of Cardiovascular and Metabolic Disorders, Duke-NUS Medical School Singapore8 College Road, 169857, Singapore, Singapore
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Seconda Università degli Studi della Campania "Luigi Vanvitelli" Napoli, Caserta, Italy
| | - Federica Cioffi
- Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, Benevento, Italy
| | - Rosalba Senese
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Seconda Università degli Studi della Campania "Luigi Vanvitelli" Napoli, Caserta, Italy
| | - Fernando Goglia
- Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, Benevento, Italy
| | - Antonia Lanni
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Seconda Università degli Studi della Campania "Luigi Vanvitelli" Napoli, Caserta, Italy.
| | - Paul M Yen
- Program of Cardiovascular and Metabolic Disorders, Duke-NUS Medical School Singapore8 College Road, 169857, Singapore, Singapore.
| | - Rohit A Sinha
- Program of Cardiovascular and Metabolic Disorders, Duke-NUS Medical School Singapore8 College Road, 169857, Singapore, Singapore.
| |
Collapse
|
166
|
Schott MB, Rasineni K, Weller SG, Schulze RJ, Sletten AC, Casey CA, McNiven MA. β-Adrenergic induction of lipolysis in hepatocytes is inhibited by ethanol exposure. J Biol Chem 2017; 292:11815-11828. [PMID: 28515323 DOI: 10.1074/jbc.m117.777748] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 05/05/2017] [Indexed: 12/21/2022] Open
Abstract
In liver steatosis (i.e. fatty liver), hepatocytes accumulate many large neutral lipid storage organelles known as lipid droplets (LDs). LDs are important in the maintenance of energy homeostasis, but the signaling mechanisms that stimulate LD metabolism in hepatocytes are poorly defined. In adipocytes, catecholamines target the β-adrenergic (β-AR)/cAMP pathway to activate cytosolic lipases and induce their recruitment to the LD surface. Therefore, the goal of this study was to determine whether hepatocytes, like adipocytes, also undergo cAMP-mediated lipolysis in response to β-AR stimulation. Using primary rat hepatocytes and human hepatoma cells, we found that treatment with the β-AR agent isoproterenol caused substantial LD loss via activation of cytosolic lipases adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL). β-Adrenergic stimulation rapidly activated PKA, which led to the phosphorylation of ATGL and HSL and their recruitment to the LD surface. To test whether this β-AR-dependent lipolysis pathway was altered in a model of alcoholic fatty liver, primary hepatocytes from rats fed a 6-week EtOH-containing Lieber-DeCarli diet were treated with cAMP agonists. Compared with controls, EtOH-exposed hepatocytes showed a drastic inhibition in β-AR/cAMP-induced LD breakdown and the phosphorylation of PKA substrates, including HSL. This observation was supported in VA-13 cells, an EtOH-metabolizing human hepatoma cell line, which displayed marked defects in both PKA activation and isoproterenol-induced ATGL translocation to the LD periphery. In summary, these findings suggest that β-AR stimulation mobilizes cytosolic lipases for LD breakdown in hepatocytes, and perturbation of this pathway could be a major consequence of chronic EtOH insult leading to fatty liver.
Collapse
Affiliation(s)
- Micah B Schott
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905
| | - Karuna Rasineni
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Shaun G Weller
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905
| | - Ryan J Schulze
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905
| | - Arthur C Sletten
- Division of Gastroenterology & Hepatology, Center for Basic Research in Digestive Diseases, Mayo Clinic, Rochester, Minnesota 55905
| | - Carol A Casey
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198; Nebraska Western Iowa Health Care System Research Service, Omaha, Nebraska 68105
| | - Mark A McNiven
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905.
| |
Collapse
|
167
|
Byun S, Kim YC, Zhang Y, Kong B, Guo G, Sadoshima J, Ma J, Kemper B, Kemper JK. A postprandial FGF19-SHP-LSD1 regulatory axis mediates epigenetic repression of hepatic autophagy. EMBO J 2017; 36:1755-1769. [PMID: 28446510 DOI: 10.15252/embj.201695500] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 03/31/2017] [Accepted: 04/04/2017] [Indexed: 12/19/2022] Open
Abstract
Lysosome-mediated autophagy is essential for cellular survival and homeostasis upon nutrient deprivation, but is repressed after feeding. Despite the emerging importance of transcriptional regulation of autophagy by nutrient-sensing factors, the role for epigenetic control is largely unexplored. Here, we show that Small Heterodimer Partner (SHP) mediates postprandial epigenetic repression of hepatic autophagy by recruiting histone demethylase LSD1 in response to a late fed-state hormone, FGF19 (hFGF19, mFGF15). FGF19 treatment or feeding inhibits macroautophagy, including lipophagy, but these effects are blunted in SHP-null mice or LSD1-depleted mice. In addition, feeding-mediated autophagy inhibition is attenuated in FGF15-null mice. Upon FGF19 treatment or feeding, SHP recruits LSD1 to CREB-bound autophagy genes, including Tfeb, resulting in dissociation of CRTC2, LSD1-mediated demethylation of gene-activation histone marks H3K4-me2/3, and subsequent accumulation of repressive histone modifications. Both FXR and SHP inhibit hepatic autophagy interdependently, but while FXR acts early, SHP acts relatively late after feeding, which effectively sustains postprandial inhibition of autophagy. This study demonstrates that the FGF19-SHP-LSD1 axis maintains homeostasis by suppressing unnecessary autophagic breakdown of cellular components, including lipids, under nutrient-rich postprandial conditions.
Collapse
Affiliation(s)
- Sangwon Byun
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Young-Chae Kim
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Yang Zhang
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Bo Kong
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Grace Guo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Jian Ma
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Byron Kemper
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jongsook Kim Kemper
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
168
|
Hahn O, Grönke S, Stubbs TM, Ficz G, Hendrich O, Krueger F, Andrews S, Zhang Q, Wakelam MJ, Beyer A, Reik W, Partridge L. Dietary restriction protects from age-associated DNA methylation and induces epigenetic reprogramming of lipid metabolism. Genome Biol 2017; 18:56. [PMID: 28351387 PMCID: PMC5370449 DOI: 10.1186/s13059-017-1187-1] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/02/2017] [Indexed: 12/11/2022] Open
Abstract
Background Dietary restriction (DR), a reduction in food intake without malnutrition, increases most aspects of health during aging and extends lifespan in diverse species, including rodents. However, the mechanisms by which DR interacts with the aging process to improve health in old age are poorly understood. DNA methylation could play an important role in mediating the effects of DR because it is sensitive to the effects of nutrition and can affect gene expression memory over time. Results Here, we profile genome-wide changes in DNA methylation, gene expression and lipidomics in response to DR and aging in female mouse liver. DR is generally strongly protective against age-related changes in DNA methylation. During aging with DR, DNA methylation becomes targeted to gene bodies and is associated with reduced gene expression, particularly of genes involved in lipid metabolism. The lipid profile of the livers of DR mice is correspondingly shifted towards lowered triglyceride content and shorter chain length of triglyceride-associated fatty acids, and these effects become more pronounced with age. Conclusions Our results indicate that DR remodels genome-wide patterns of DNA methylation so that age-related changes are profoundly delayed, while changes at loci involved in lipid metabolism affect gene expression and the resulting lipid profile. Electronic supplementary material The online version of this article (doi:10.1186/s13059-017-1187-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Oliver Hahn
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany.,Cellular Networks and Systems Biology, CECAD, University of Cologne, Joseph-Stelzmann-Str. 26, Cologne, 50931, Germany
| | - Sebastian Grönke
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Thomas M Stubbs
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK
| | - Gabriella Ficz
- Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Oliver Hendrich
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Felix Krueger
- Bioinformatics Group, The Babraham Institute, Cambridge, CB22 3AT, UK
| | - Simon Andrews
- Bioinformatics Group, The Babraham Institute, Cambridge, CB22 3AT, UK
| | - Qifeng Zhang
- Inositide Lab, The Babraham Institute, Cambridge, CB22 3AT, UK
| | | | - Andreas Beyer
- Cellular Networks and Systems Biology, CECAD, University of Cologne, Joseph-Stelzmann-Str. 26, Cologne, 50931, Germany. .,Center for Molecular Medicine Cologne, University of Cologne, Cologne, 50931, Germany.
| | - Wolf Reik
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK. .,The Wellcome Trust Sanger Institute, Cambridge, CB10 1SA, UK.
| | - Linda Partridge
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany. .,Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
169
|
Yang G, Wang Y, Feng J, Liu Y, Wang T, Zhao M, Ye L, Zhang X. Aspirin suppresses the abnormal lipid metabolism in liver cancer cells via disrupting an NFκB-ACSL1 signaling. Biochem Biophys Res Commun 2017; 486:827-832. [PMID: 28359761 DOI: 10.1016/j.bbrc.2017.03.139] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 03/26/2017] [Indexed: 01/04/2023]
Abstract
Abnormal lipid metabolism is a hallmark of tumorigenesis. Hence, the alterations of metabolism enhance the development of hepatocellular carcinoma (HCC). Aspirin is able to inhibit the growth of cancers through targeting nuclear factor κB (NF-κB). However, the role of aspirin in disrupting abnormal lipid metabolism in HCC remains poorly understood. In this study, we report that aspirin can suppress the abnormal lipid metabolism of HCC cells through inhibiting acyl-CoA synthetase long-chain family member 1 (ACSL1), a lipid metabolism-related enzyme. Interestingly, oil red O staining showed that aspirin suppressed lipogenesis in HepG2 cells and Huh7 cells in a dose-dependent manner. In addition, aspirin attenuated the levels of triglyceride and cholesterol in the cells, respectively. Strikingly, we identified that aspirin was able to down-regulate ACSL1 at the levels of mRNA and protein. Moreover, we validated that aspirin decreased the nuclear levels of NF-κB in HepG2 cells. Mechanically, PDTC, an inhibitor of NF-κB, could down-regulate ACSL1 at the levels of mRNA and protein in the cells. Functionally, PDTC reduced the levels of lipid droplets, triglyceride and cholesterol in HepG2 cells. Thus, we conclude that aspirin suppresses the abnormal lipid metabolism in HCC cells via disrupting an NFκB-ACSL1 signaling. Our finding provides new insights into the mechanism by which aspirin inhibits abnormal lipid metabolism of HCC. Therapeutically, aspirin is potentially available for HCC through controlling abnormal lipid metabolism.
Collapse
Affiliation(s)
- Guang Yang
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yuan Wang
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jinyan Feng
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yunxia Liu
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Tianjiao Wang
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Man Zhao
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Lihong Ye
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaodong Zhang
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
170
|
Gemmink A, Bakker LEH, Guigas B, Kornips E, Schaart G, Meinders AE, Jazet IM, Hesselink MKC. Lipid droplet dynamics and insulin sensitivity upon a 5-day high-fat diet in Caucasians and South Asians. Sci Rep 2017; 7:42393. [PMID: 28195217 PMCID: PMC5307956 DOI: 10.1038/srep42393] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 01/09/2017] [Indexed: 12/25/2022] Open
Abstract
A 5-day High-Fat High-Calorie diet (HFHC-diet) reduces insulin-stimulated glucose disposal (Rd) in South Asian, but not Caucasian healthy lean males. We aimed to investigate if differences in myocellular lipid handling are underlying this differential response. A two-step hyperinsulinemic-euglycemic clamp and muscle biopsies were performed in 12 healthy lean Caucasian and South Asian males (BMI < 25 kg/m2, 19-25 years) before and after a 5-day HFHC-diet (regular diet + 375 mL cream/day; 1275 kcal/day; 94% fat). Triglyceride extractions and Western Blots for lipid droplet and mitochondrial proteins were performed. Intramyocellular lipid content and HFHC-diet response were similar between ethnicities (group effect: P = 0.094; diet effect: +~30%, P = 0.044). PLIN5 protein content increased upon the HFHC-diet (P = 0.031) and tended to be higher in South Asians (0.87 ± 0.42 AU vs. 1.35 ± 0.58 AU, P = 0.07). 4-HNE tended to increase in South Asians upon the HFHC-diet (interaction effect: P = 0.057). In Caucasians ΔPLIN5 content correlated with ΔRd (Caucasians: r = 0.756, P = 0.011; South Asians: r = -0.085, P = 0.816), while in South Asians Δ4-HNE associated with ΔPLIN5 content (Caucasians: r = 0.312, P = 0.380; South Asians: r = 0.771, P = 0.003). These data indicate that in Caucasians, PLIN5 may be protective against HFHC-diet induced insulin resistance, which for reasons not yet understood is not observed in South Asians, who possess increased lipid peroxidation levels.
Collapse
Affiliation(s)
- Anne Gemmink
- Department of Human Biology and Human Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6200 MD, Maastricht, The Netherlands
| | - Leontine E H Bakker
- Department of Internal Medicine, section Endocrinology, Leiden University Medical Center, The Netherlands
| | - Bruno Guigas
- Department of Molecular Cell Biology, Leiden University Medical Center, The Netherlands.,Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Esther Kornips
- Department of Human Biology and Human Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6200 MD, Maastricht, The Netherlands
| | - Gert Schaart
- Department of Human Biology and Human Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6200 MD, Maastricht, The Netherlands
| | - A Edo Meinders
- Department of Internal Medicine, section Endocrinology, Leiden University Medical Center, The Netherlands
| | - Ingrid M Jazet
- Department of Internal Medicine, section Endocrinology, Leiden University Medical Center, The Netherlands
| | - Matthijs K C Hesselink
- Department of Human Biology and Human Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6200 MD, Maastricht, The Netherlands
| |
Collapse
|
171
|
Irshad Z, Dimitri F, Christian M, Zammit VA. Diacylglycerol acyltransferase 2 links glucose utilization to fatty acid oxidation in the brown adipocytes. J Lipid Res 2017; 58:15-30. [PMID: 27836993 PMCID: PMC5234708 DOI: 10.1194/jlr.m068197] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 10/18/2016] [Indexed: 01/03/2023] Open
Abstract
Brown adipose tissue uptake of glucose and fatty acids is very high during nonshivering thermogenesis. Adrenergic stimulation markedly increases glucose uptake, de novo lipogenesis, and FA oxidation simultaneously. The mechanism that enables this concerted response has hitherto been unknown. Here, we find that in primary brown adipocytes and brown adipocyte-derived cell line (IMBAT-1), acute inhibition and longer-term knockdown of DGAT2 links the increased de novo synthesis of fatty acids from glucose to a pool of TAG that is simultaneously hydrolyzed, providing FA for mitochondrial oxidation. DGAT1 does not contribute to this pathway, but uses exogenous FA and glycerol to synthesize a functionally distinct pool of TAG to which DGAT2 also contributes. The DGAT2-dependent channelling of 14C from glucose into TAG and CO2 was reproduced in β3-agonist-stimulated primary brown adipocytes. Knockdown of DGAT2 in IMBAT-1 affected the mRNA levels of UCP1 and genes important in FA activation and esterification. Therefore, in β3-agonist activated brown adipocytes, DGAT2 specifically enables channelling of de novo synthesized FA into a rapidly mobilized pool of TAG, which is simultaneously hydrolyzed to provide substrates for mitochondrial fatty acid oxidation.
Collapse
Affiliation(s)
- Zehra Irshad
- Translational and Experimental Medicine, Division of Biomedical Sciences, Warwick Medical School, CV4 7AL, United Kingdom
| | - Federica Dimitri
- Translational and Experimental Medicine, Division of Biomedical Sciences, Warwick Medical School, CV4 7AL, United Kingdom
| | - Mark Christian
- Translational and Experimental Medicine, Division of Biomedical Sciences, Warwick Medical School, CV4 7AL, United Kingdom
| | - Victor A Zammit
- Translational and Experimental Medicine, Division of Biomedical Sciences, Warwick Medical School, CV4 7AL, United Kingdom
| |
Collapse
|
172
|
Ghosh M, Niyogi S, Bhattacharyya M, Adak M, Nayak DK, Chakrabarti S, Chakrabarti P. Ubiquitin Ligase COP1 Controls Hepatic Fat Metabolism by Targeting ATGL for Degradation. Diabetes 2016; 65:3561-3572. [PMID: 27658392 DOI: 10.2337/db16-0506] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 09/14/2016] [Indexed: 11/13/2022]
Abstract
Optimal control of hepatic lipid metabolism is critical for organismal metabolic fitness. In liver, adipose triglyceride lipase (ATGL) serves as a major triacylglycerol (TAG) lipase and controls the bulk of intracellular lipid turnover. However, regulation of ATGL expression and its functional implications in hepatic lipid metabolism, particularly in the context of fatty liver disease, is unclear. We show that E3 ubiquitin ligase COP1 (also known as RFWD2) binds to the consensus VP motif of ATGL and targets it for proteasomal degradation by K-48 linked polyubiquitination, predominantly at the lysine 100 residue. COP1 thus serves as a critical regulator of hepatocyte TAG content, fatty acid mobilization, and oxidation. Moreover, COP1-mediated regulation of hepatic lipid metabolism requires optimum ATGL expression for its metabolic outcome. In vivo, adenovirus-mediated depletion of COP1 ameliorates high-fat diet-induced steatosis in mouse liver and improves liver function. Our study thus provides new insights into the regulation of hepatic lipid metabolism by the ubiquitin-proteasome system and suggests COP1 as a potential therapeutic target for nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Mainak Ghosh
- Division of Cell Biology and Physiology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Sougata Niyogi
- Division of Cell Biology and Physiology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Madhumita Bhattacharyya
- Division of Structural Biology and Bioinformatics, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Moumita Adak
- Division of Cell Biology and Physiology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Dipak K Nayak
- Nuclear Medicine Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Saikat Chakrabarti
- Division of Structural Biology and Bioinformatics, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Partha Chakrabarti
- Division of Cell Biology and Physiology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
173
|
Chen G, Yu D, Nian X, Liu J, Koenig RJ, Xu B, Sheng L. LncRNA SRA promotes hepatic steatosis through repressing the expression of adipose triglyceride lipase (ATGL). Sci Rep 2016; 6:35531. [PMID: 27759039 PMCID: PMC5069493 DOI: 10.1038/srep35531] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 09/30/2016] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), the most common form of chronic liver disease, manifests as an over-accumulation of hepatic fat. We have recently shown that mice with genetic knockout of a long non-coding RNA (lncRNA) steroid receptor RNA activator (SRA) (SRAKO) are resistant to high fat diet-induced obesity with a phenotype that includes improved glucose tolerance and attenuated hepatic steatosis. The underlying mechanism was investigated in the present study. We found that hepatic levels of SRA and adipose triglyceride lipase (ATGL), a major hepatic triacylglycerol (TAG) hydrolase, were inversely regulated by fasting in mice, and the expression of liver ATGL was induced by SRAKO under normal and high fat diet (HFD) feeding. Loss of SRA in primary hepatocytes or a hepatocyte cell line upregulates, but forced expression of SRA inhibits ATGL expression and free fatty acids (FFA) β-oxidation. SRA inhibits ATGL promoter activity, primarily by inhibiting the otherwise-inductive effects of the transcription factor, forkhead box protein O1 (FoxO1). Our data reveal a novel function of SRA in promoting hepatic steatosis through repression of ATGL expression.
Collapse
Affiliation(s)
- Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Dongsheng Yu
- Department of Pharmacology, School of Basic Medical Science, Nanjing Medical University, 140 Hanzhong Rd., Nanjing, Jiangsu, 210029, China
| | - Xue Nian
- Department of Pharmacology, School of Basic Medical Science, Nanjing Medical University, 140 Hanzhong Rd., Nanjing, Jiangsu, 210029, China
| | - Junyi Liu
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ronald J Koenig
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, MI 48109-5678, USA
| | - Bin Xu
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, MI 48109-5678, USA
| | - Liang Sheng
- Department of Pharmacology, School of Basic Medical Science, Nanjing Medical University, 140 Hanzhong Rd., Nanjing, Jiangsu, 210029, China
| |
Collapse
|
174
|
Polizzi A, Fouché E, Ducheix S, Lasserre F, Marmugi AP, Mselli-Lakhal L, Loiseau N, Wahli W, Guillou H, Montagner A. Hepatic Fasting-Induced PPARα Activity Does Not Depend on Essential Fatty Acids. Int J Mol Sci 2016; 17:ijms17101624. [PMID: 27669233 PMCID: PMC5085657 DOI: 10.3390/ijms17101624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/05/2016] [Accepted: 09/15/2016] [Indexed: 12/13/2022] Open
Abstract
The liver plays a central role in the regulation of fatty acid metabolism, which is highly sensitive to transcriptional responses to nutrients and hormones. Transcription factors involved in this process include nuclear hormone receptors. One such receptor, PPARα, which is highly expressed in the liver and activated by a variety of fatty acids, is a critical regulator of hepatic fatty acid catabolism during fasting. The present study compared the influence of dietary fatty acids and fasting on hepatic PPARα-dependent responses. Pparα−/− male mice and their wild-type controls were fed diets containing different fatty acids for 10 weeks prior to being subjected to fasting or normal feeding. In line with the role of PPARα in sensing dietary fatty acids, changes in chronic dietary fat consumption influenced liver damage during fasting. The changes were particularly marked in mice fed diets lacking essential fatty acids. However, fasting, rather than specific dietary fatty acids, induced acute PPARα activity in the liver. Taken together, the data imply that the potent signalling involved in triggering PPARα activity during fasting does not rely on essential fatty acid-derived ligand.
Collapse
Affiliation(s)
- Arnaud Polizzi
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
| | - Edwin Fouché
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
| | - Simon Ducheix
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
| | - Frédéric Lasserre
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
| | - Alice P Marmugi
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
| | | | - Nicolas Loiseau
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
| | - Walter Wahli
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
- Lee Kong Chian School of Medicine, Nanyang Technological University, 637553 Singapore, Singapore.
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Hervé Guillou
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
| | | |
Collapse
|
175
|
Rodrigues AH, Moreira CCL, Mario ÉG, de Souza Cordeiro LM, Avelar GF, Botion LM, Chaves VE. Differential modulation of cytosolic lipases activities in liver and adipose tissue by high-carbohydrate diets. Endocrine 2016; 53:423-32. [PMID: 26874528 DOI: 10.1007/s12020-016-0886-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 02/02/2016] [Indexed: 10/22/2022]
Abstract
Several studies have demonstrated that a high-fructose (FRUC) diet induces metabolic and haemodynamic abnormalities, known as the metabolic syndrome, which are characterised by obesity, glucose intolerance, insulin resistance, dyslipidaemia and hypertension. In this study, the effect of a FRUC diet (60 % fructose) for 8 weeks on the metabolism of lipids in liver and epididymal adipose tissue from Wistar rats was compared with the AIN-93M diet and the effects of the AIN-93M diet were compared with a chow diet. The FRUC diet induced marked increases in both hepatocyte lipid droplet volume and postprandial serum levels of triacylglycerol (TAG), but reduced the postprandial serum levels of insulin. The AIN-93M diet induced marked increases in the hepatocyte lipid droplet volume and the serum levels of insulin, without affecting the serum levels of TAG. We found that isocaloric substitution of cornstarch, dextrinised cornstarch and sucrose (AIN-93M diet) for fructose did not affect the hepatic VLDL-TAG secretion and adipose tissue glucose uptake, lipolysis and cytosolic lipases activities in rats. However, the high-fructose diet induced a severe steatosis in liver accompanied by a decrease in cytosolic lipases activities. In adipose tissue, the FRUC diet induced a decrease in the lipoprotein lipase activity, and an increase in lipogenesis. FRUC and AIN-93M diets induced changes in lipid homeostasis in liver and adipose tissue by distinct biochemical mechanisms.
Collapse
Affiliation(s)
| | - Carolina Campos Lima Moreira
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Érica Guilhen Mario
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Letícia Maria de Souza Cordeiro
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gleide Fernandes Avelar
- Department of Morphology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Leida Maria Botion
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Valéria Ernestânia Chaves
- Laboratory of Physiology, Federal University of São João del-Rei, Avenida Sebastião Gonçalves Coelho, 400, Chanadour, Divinópolis, Minas Gerais, 35501-296, Brazil.
| |
Collapse
|
176
|
Sun J, Ji H, Li XX, Shi XC, Du ZY, Chen LQ. Lipolytic enzymes involving lipolysis in Teleost: Synteny, structure, tissue distribution, and expression in grass carp (Ctenopharyngodon idella). Comp Biochem Physiol B Biochem Mol Biol 2016; 198:110-8. [DOI: 10.1016/j.cbpb.2016.04.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 04/25/2016] [Accepted: 04/25/2016] [Indexed: 02/06/2023]
|
177
|
AMPK Phosphorylates Desnutrin/ATGL and Hormone-Sensitive Lipase To Regulate Lipolysis and Fatty Acid Oxidation within Adipose Tissue. Mol Cell Biol 2016; 36:1961-76. [PMID: 27185873 DOI: 10.1128/mcb.00244-16] [Citation(s) in RCA: 216] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 05/06/2016] [Indexed: 01/10/2023] Open
Abstract
The role of AMP-activated protein kinase (AMPK) in promoting fatty acid (FA) oxidation in various tissues, such as liver and muscle, has been well understood. However, the role of AMPK in lipolysis and FA metabolism in adipose tissue has been controversial. To investigate the role of AMPK in the regulation of adipose lipolysis in vivo, we generated mice with adipose-tissue-specific knockout of both the α1 and α2 catalytic subunits of AMPK (AMPK-ASKO mice) by using aP2-Cre and adiponectin-Cre. Both models of AMPK-ASKO ablation show no changes in desnutrin/ATGL levels but have defective phosphorylation of desnutrin/ATGL at S406 to decrease its triacylglycerol (TAG) hydrolase activity, lowering basal lipolysis in adipose tissue. These mice also show defective phosphorylation of hormone-sensitive lipase (HSL) at S565, with higher phosphorylation at protein kinase A sites S563 and S660, increasing its hydrolase activity and isoproterenol-stimulated lipolysis. With higher overall adipose lipolysis, both models of AMPK-ASKO mice are lean, having smaller adipocytes with lower TAG and higher intracellular free-FA levels. Moreover, FAs from higher lipolysis activate peroxisome proliferator-activated receptor delta to induce FA oxidative genes and increase FA oxidation and energy expenditure. Overall, for the first time, we provide in vivo evidence of the role of AMPK in the phosphorylation and regulation of desnutrin/ATGL and HSL and thus adipose lipolysis.
Collapse
|
178
|
Zhu S, Park S, Lim Y, Shin S, Han SN. Korean pine nut oil replacement decreases intestinal lipid uptake while improves hepatic lipid metabolism in mice. Nutr Res Pract 2016; 10:477-486. [PMID: 27698954 PMCID: PMC5037064 DOI: 10.4162/nrp.2016.10.5.477] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 03/28/2016] [Accepted: 05/17/2016] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND/OBJECTIVES Consumption of pine nut oil (PNO) was shown to reduce weight gain and attenuate hepatic steatosis in mice fed a high-fat diet (HFD). The aim of this study was to examine the effects of PNO on both intestinal and hepatic lipid metabolism in mice fed control or HFD. MATERIALS/METHODS Five-week-old C57BL/6 mice were fed control diets containing 10% energy fat from either Soybean Oil (SBO) or PNO, or HFD containing 15% energy fat from lard and 30% energy fat from SBO or PNO for 12 weeks. Expression of genes related to intestinal fatty acid (FA) uptake and channeling (Cd36, Fatp4, Acsl5, Acbp), intestinal chylomicron synthesis (Mtp, ApoB48, ApoA4), hepatic lipid uptake and channeling (Lrp1, Fatp5, Acsl1, Acbp), hepatic triacylglycerol (TAG) lipolysis and FA oxidation (Atgl, Cpt1a, Acadl, Ehhadh, Acaa1), as well as very low-density lipoprotein (VLDL) assembly (ApoB100) were determined by real-time PCR. RESULTS In intestine, significantly lower Cd36 mRNA expression (P < 0.05) and a tendency of lower ApoA4 mRNA levels (P = 0.07) was observed in PNO-fed mice, indicating that PNO consumption may decrease intestinal FA uptake and chylomicron assembly. PNO consumption tended to result in higher hepatic mRNA levels of Atgl (P = 0.08) and Cpt1a (P = 0.05). Significantly higher hepatic mRNA levels of Acadl and ApoB100 were detected in mice fed PNO diet (P < 0.05). These results suggest that PNO could increase hepatic TAG metabolism; mitochondrial fatty acid oxidation and VLDL assembly. CONCLUSIONS PNO replacement in the diet might function in prevention of excessive lipid uptake by intestine and improve hepatic lipid metabolism in both control diet and HFD fed mice.
Collapse
Affiliation(s)
- Shuang Zhu
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Soyoung Park
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Yeseo Lim
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Sunhye Shin
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Sung Nim Han
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea.; Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
179
|
Li Y, Zalzala M, Jadhav K, Xu Y, Kasumov T, Yin L, Zhang Y. Carboxylesterase 2 prevents liver steatosis by modulating lipolysis, endoplasmic reticulum stress, and lipogenesis and is regulated by hepatocyte nuclear factor 4 alpha in mice. Hepatology 2016; 63:1860-74. [PMID: 26806650 PMCID: PMC4874867 DOI: 10.1002/hep.28472] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/15/2016] [Accepted: 01/20/2016] [Indexed: 12/27/2022]
Abstract
UNLABELLED Nonalcoholic fatty liver disease (NAFLD) is a common liver disease that ranges from simple steatosis to nonalcoholic steatohepatitis (NASH). So far, the underlying mechanism remains poorly understood. Here, we show that hepatic carboxylesterase 2 (CES2) is markedly reduced in NASH patients, diabetic db/db mice, and high-fat diet (HFD)-fed mice. Restoration of hepatic CES2 expression in db/db or HFD-fed mice markedly ameliorates liver steatosis and insulin resistance. In contrast, knockdown of hepatic CES2 causes liver steatosis and damage in chow- or Western diet-fed C57BL/6 mice. Mechanistically, we demonstrate that CES2 has triglyceride hydrolase activity. As a result, gain of hepatic CES2 function increases fatty acid oxidation and inhibits lipogenesis, whereas loss of hepatic CES2 stimulates lipogenesis by inducing endoplasmic reticulum stress. We further show that loss of hepatic CES2 stimulates lipogenesis in a sterol regulatory element-binding protein 1 (SREBP-1)-dependent manner. Finally, we show that hepatocyte nuclear factor 4 alpha (HNF-4α) plays a key role in controlling hepatic CES2 expression in diabetes, obesity, or NASH. CONCLUSION CES2 plays a protective role in development of NAFLD. Targeting the HNF-4α/CES2 pathway may be useful for treatment of NAFLD. (Hepatology 2016;63:1860-1874).
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Munaf Zalzala
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA,Department of Pharmacology and toxicology, College of Pharmacy, University of Baghdad, Baghdad, Iraq
| | - Kavita Jadhav
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Yang Xu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Takhar Kasumov
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Liya Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Yanqiao Zhang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| |
Collapse
|
180
|
van der Veen JN, Lingrell S, Gao X, Quiroga AD, Takawale A, Armstrong EA, Yager JY, Kassiri Z, Lehner R, Vance DE, Jacobs RL. Pioglitazone attenuates hepatic inflammation and fibrosis in phosphatidylethanolamine N-methyltransferase-deficient mice. Am J Physiol Gastrointest Liver Physiol 2016; 310:G526-38. [PMID: 26797396 DOI: 10.1152/ajpgi.00243.2015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 12/21/2015] [Indexed: 01/31/2023]
Abstract
Phosphatidylethanolamine N-methyltransferase (PEMT) is an important enzyme in hepatic phosphatidylcholine (PC) biosynthesis. Pemt(-/-) mice are protected against high-fat diet (HFD)-induced obesity and insulin resistance; however, these mice develop nonalcoholic fatty liver disease (NAFLD). We hypothesized that peroxisomal proliferator-activated receptor-γ (PPARγ) activation by pioglitazone might stimulate adipocyte proliferation, thereby directing lipids from the liver toward white adipose tissue. Pioglitazone might also act directly on PPARγ in the liver to improve NAFLD. Pemt(+/+) and Pemt(-/-) mice were fed a HFD with or without pioglitazone (20 mg·kg(-1)·day(-1)) for 10 wk. Pemt(-/-) mice were protected from HFD-induced obesity but developed NAFLD. Treatment with pioglitazone caused an increase in body weight gain in Pemt(-/-) mice that was mainly due to increased adiposity. Moreover, pioglitazone improved NAFLD in Pemt(-/-) mice, as indicated by a 35% reduction in liver weight and a 57% decrease in plasma alanine transaminase levels. Livers from HFD-fed Pemt(-/-) mice were steatotic, inflamed, and fibrotic. Hepatic steatosis was still evident in pioglitazone-treated Pemt(-/-) mice; however, treatment with pioglitazone reduced hepatic fibrosis, as evidenced by reduced Sirius red staining and lowered mRNA levels of collagen type Iα1 (Col1a1), tissue inhibitor of metalloproteinases 1 (Timp1), α-smooth muscle actin (Acta2), and transforming growth factor-β (Tgf-β). Similarly, oxidative stress and inflammation were reduced in livers from Pemt(-/-) mice upon treatment with pioglitazone. Together, these data show that activation of PPARγ in HFD-fed Pemt(-/-) mice improved liver function, while these mice were still protected against diet-induced obesity and insulin resistance.
Collapse
Affiliation(s)
- Jelske N van der Veen
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada; Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Susanne Lingrell
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada; Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Xia Gao
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada; Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ariel D Quiroga
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Abhijit Takawale
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; and
| | - Edward A Armstrong
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Jerome Y Yager
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; and
| | - Richard Lehner
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Dennis E Vance
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada; Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - René L Jacobs
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada; Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
181
|
Zhang W, Bu SY, Mashek MT, O-Sullivan I, Sibai Z, Khan SA, Ilkayeva O, Newgard CB, Mashek DG, Unterman TG. Integrated Regulation of Hepatic Lipid and Glucose Metabolism by Adipose Triacylglycerol Lipase and FoxO Proteins. Cell Rep 2016; 15:349-59. [PMID: 27050511 DOI: 10.1016/j.celrep.2016.03.021] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 02/23/2016] [Accepted: 03/03/2016] [Indexed: 12/16/2022] Open
Abstract
Metabolism is a highly integrated process that is coordinately regulated between tissues and within individual cells. FoxO proteins are major targets of insulin action and contribute to the regulation of gluconeogenesis, glycolysis, and lipogenesis in the liver. However, the mechanisms by which FoxO proteins exert these diverse effects in an integrated fashion remain poorly understood. We report that FoxO proteins also exert important effects on intrahepatic lipolysis and fatty acid oxidation via the regulation of adipose triacylglycerol lipase (ATGL), which mediates the first step in lipolysis, and its inhibitor, the G0/S1 switch 2 gene (G0S2). We also find that ATGL-dependent lipolysis plays a critical role in mediating diverse effects of FoxO proteins in the liver, including effects on gluconeogenic, glycolytic, and lipogenic gene expression and metabolism. These results indicate that intrahepatic lipolysis plays a critical role in mediating and integrating the regulation of glucose and lipid metabolism downstream of FoxO proteins.
Collapse
Affiliation(s)
- Wenwei Zhang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Medical Research Service, Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - So Young Bu
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mara T Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - InSug O-Sullivan
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Medical Research Service, Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Zakaria Sibai
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Medical Research Service, Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Salmaan A Khan
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC 27710, USA; Department of Pharmacology, Duke University, Durham, NC 27710, USA; Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC 27710, USA; Department of Pharmacology, Duke University, Durham, NC 27710, USA; Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Terry G Unterman
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Medical Research Service, Jesse Brown VA Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
182
|
Abstract
Lipid droplets are the intracellular sites for neutral lipid storage. They are critical for lipid metabolism and energy homeostasis, and their dysfunction has been linked to many diseases. Accumulating evidence suggests that the roles lipid droplets play in biology are significantly broader than previously anticipated. Lipid droplets are the source of molecules important in the nucleus: they can sequester transcription factors and chromatin components and generate the lipid ligands for certain nuclear receptors. Lipid droplets have also emerged as important nodes for fatty acid trafficking, both inside the cell and between cells. In immunity, new roles for droplets, not directly linked to lipid metabolism, have been uncovered, with evidence that they act as assembly platforms for specific viruses and as reservoirs for proteins that fight intracellular pathogens. Until recently, knowledge about droplets in the nervous system has been minimal, but now there are multiple links between lipid droplets and neurodegeneration: many candidate genes for hereditary spastic paraplegia also have central roles in lipid-droplet formation and maintenance, and mitochondrial dysfunction in neurons can lead to transient accumulation of lipid droplets in neighboring glial cells, an event that may, in turn, contribute to neuronal damage. As the cell biology and biochemistry of lipid droplets become increasingly well understood, the next few years should yield many new mechanistic insights into these novel functions of lipid droplets.
Collapse
|
183
|
Ress C, Kaser S. Mechanisms of intrahepatic triglyceride accumulation. World J Gastroenterol 2016; 22:1664-1673. [PMID: 26819531 PMCID: PMC4721997 DOI: 10.3748/wjg.v22.i4.1664] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/20/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatic steatosis defined as lipid accumulation in hepatocytes is very frequently found in adults and obese adolescents in the Western World. Etiologically, obesity and associated insulin resistance or excess alcohol intake are the most frequent causes of hepatic steatosis. However, steatosis also often occurs with chronic hepatitis C virus (HCV) infection and is also found in rare but potentially life-threatening liver diseases of pregnancy. Clinical significance and outcome of hepatic triglyceride accumulation are highly dependent on etiology and histological pattern of steatosis. This review summarizes current concepts of pathophysiology of common causes of hepatic steatosis, including non-alcoholic fatty liver disease (NAFLD), alcoholic fatty liver disease, chronic HCV infections, drug-induced forms of hepatic steatosis, and acute fatty liver of pregnancy. Regarding the pathophysiology of NAFLD, this work focuses on the close correlation between insulin resistance and hepatic triglyceride accumulation, highlighting the potential harmful effects of systemic insulin resistance on hepatic metabolism of fatty acids on the one side and the role of lipid intermediates on insulin signalling on the other side. Current studies on lipid droplet morphogenesis have identified novel candidate proteins and enzymes in NAFLD.
Collapse
|
184
|
Grasselli E, Voci A, Demori I, Vecchione G, Compalati AD, Gallo G, Goglia F, De Matteis R, Silvestri E, Vergani L. Triglyceride Mobilization from Lipid Droplets Sustains the Anti-Steatotic Action of Iodothyronines in Cultured Rat Hepatocytes. Front Physiol 2016; 6:418. [PMID: 26793120 PMCID: PMC4709507 DOI: 10.3389/fphys.2015.00418] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 12/21/2015] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue, dietary lipids and de novo lipogenesis are sources of hepatic free fatty acids (FFAs) that are stored in lipid droplets (LDs) as triacylglycerols (TAGs). Destiny of TAGs stored in LDs is determined by LD proteomic equipment. When adipose triglyceride lipase (ATGL) localizes at LD surface the lipid mobilization is stimulated. In this work, an in vitro model of cultured rat hepatocytes mimicking a mild steatosis condition was used to investigate the direct lipid-lowering action of iodothyronines, by focusing, in particular, on LD-associated proteins, FFA oxidation and lipid secretion. Our results demonstrate that in “steatotic” hepatocytes iodothyronines reduced the lipid excess through the recruitment of ATGL on LD surface, and the modulation of the LD-associated proteins Rab18 and TIP47. As an effect of ATGL recruitment, iodothyronines stimulated the lipid mobilization from LDs then followed by the up-regulation of carnitine-palmitoyl-transferase (CPT1) expression and the stimulation of cytochrome-c oxidase (COX) activity that seems to indicate a stimulation of mitochondrial function. The lipid lowering action of iodothyronines did not depend on increased TAG secretion. On the basis of our data, ATGL could be indicated as an early mediator of the lipid-lowering action of iodothyronines able to channel hydrolyzed FFAs toward mitochondrial beta-oxidation rather than secretion.
Collapse
Affiliation(s)
- Elena Grasselli
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di GenovaGenova, Italia; Istituto Nazionale Biostrutture e BiosistemiRoma, Italia
| | - Adriana Voci
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova Genova, Italia
| | - Ilaria Demori
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova Genova, Italia
| | - Giulia Vecchione
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova Genova, Italia
| | - Andrea D Compalati
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova Genova, Italia
| | - Gabriella Gallo
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova Genova, Italia
| | - Fernando Goglia
- Dipartimento di Scienze e Tecnologie, Università del Sannio Benevento, Italia
| | - Rita De Matteis
- Dipartimento di Scienze Biomolecolari, Università di Urbino Urbino, Italia
| | - Elena Silvestri
- Dipartimento di Scienze e Tecnologie, Università del Sannio Benevento, Italia
| | - Laura Vergani
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di GenovaGenova, Italia; Istituto Nazionale Biostrutture e BiosistemiRoma, Italia
| |
Collapse
|
185
|
Urrutia RA, Kalinec F. Biology and pathobiology of lipid droplets and their potential role in the protection of the organ of Corti. Hear Res 2015; 330:26-38. [PMID: 25987503 PMCID: PMC5391798 DOI: 10.1016/j.heares.2015.04.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/17/2015] [Accepted: 04/21/2015] [Indexed: 12/20/2022]
Abstract
The current review article seeks to extend our understanding on the role of lipid droplets within the organ of Corti. In addition to presenting an overview of the current information about the origin, structure and function of lipid droplets we draw inferences from the collective body of knowledge about this cellular organelle to build a conceptual framework to better understanding their role in auditory function. This conceptual model considers that lipid droplets play a significant role in the synthesis, storage, and release of lipids and proteins for energetic use and/or modulating cell signaling pathways. We describe the role and mechanism by which LD play a role in human diseases, and we also review emerging data from our laboratory revealing the potential role of lipid droplets from Hensen cells in the auditory organ. We suggest that lipid droplets might help to develop rapidly and efficiently the resolution phase of inflammatory responses in the mammalian cochlea, preventing inflammatory damage of the delicate inner ear structures and, consequently, sensorineural hearing loss.
Collapse
Affiliation(s)
- Raul A Urrutia
- Epigenetics and Chromatin Dynamics Laboratory, Translational Epigenomic Program, Center for Individualized Medicine (CIM) Mayo Clinic, Rochester, MN 55905, USA
| | - Federico Kalinec
- Laboratory of Auditory Cell Biology, Department of Head & Neck Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
186
|
Suciu M, Gruia AT, Nica DV, Azghadi SM, Mic AA, Mic FA. Acetaminophen-induced liver injury: Implications for temporal homeostasis of lipid metabolism and eicosanoid signaling pathway. Chem Biol Interact 2015; 242:335-44. [DOI: 10.1016/j.cbi.2015.10.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/29/2015] [Accepted: 10/23/2015] [Indexed: 12/26/2022]
|
187
|
Andrés-Blasco I, Herrero-Cervera A, Vinué Á, Martínez-Hervás S, Piqueras L, Sanz MJ, Burks DJ, González-Navarro H. Hepatic lipase deficiency produces glucose intolerance, inflammation and hepatic steatosis. J Endocrinol 2015; 227:179-91. [PMID: 26423094 DOI: 10.1530/joe-15-0219] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/30/2015] [Indexed: 12/15/2022]
Abstract
Metabolic syndrome and type 2 diabetes mellitus constitute a major problem to global health, and their incidence is increasing at an alarming rate. Non-alcoholic fatty liver disease, which affects up to 90% of obese people and nearly 70% of the overweight, is commonly associated with MetS characteristics such as obesity, insulin resistance, hypertension and dyslipidemia. In the present study, we demonstrate that hepatic lipase (HL)-inactivation in mice fed with a high-fat, high-cholesterol diet produced dyslipidemia including hypercholesterolemia, hypertriglyceridemia and increased non-esterified fatty acid levels. These changes were accompanied by glucose intolerance, pancreatic and hepatic inflammation and steatosis. In addition, compared with WT mice, HL(-/-) mice exhibited enhanced circulating MCP1 levels, monocytosis and higher percentage of CD4+Th17+ cells. Consistent with increased inflammation, livers from HL(-/-) mice had augmented activation of the stress SAPK/JNK- and p38-pathways compared with the activation levels of the kinases in livers from WT mice. Analysis of HL(-/-) and WT mice fed regular chow diet showed dyslipidemia and glucose intolerance in HL(-/-) mice without any other changes in inflammation or hepatic steatosis. Altogether, these results indicate that dyslipidemia induced by HL-deficiency in combination with a high-fat, high-cholesterol diet promotes hepatic steatosis and inflammation in mice which are, at least in part, mediated by the activation of the stress SAPK/JNK- and p38-pathways. Future studies are warranted to asses the viability of therapeutic strategies based on the modulation of these kinases to reduce hepatic steatosis associated to lipase dysfunction.
Collapse
Affiliation(s)
- Irene Andrés-Blasco
- Institute of Health Research-INCLIVAAvenida Menéndez Pelayo, 4, 46010 Valencia, SpainEndocrinology and Nutrition Department Clinic Hospital and Department of MedicineUniversity of Valencia, Valencia, SpainCIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM)Valencia, SpainDepartment of FarmacologyUniversity of Valencia, Valencia, SpainCentro de Investigación Príncipe FelipeValencia, Spain
| | - Andrea Herrero-Cervera
- Institute of Health Research-INCLIVAAvenida Menéndez Pelayo, 4, 46010 Valencia, SpainEndocrinology and Nutrition Department Clinic Hospital and Department of MedicineUniversity of Valencia, Valencia, SpainCIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM)Valencia, SpainDepartment of FarmacologyUniversity of Valencia, Valencia, SpainCentro de Investigación Príncipe FelipeValencia, Spain
| | - Ángela Vinué
- Institute of Health Research-INCLIVAAvenida Menéndez Pelayo, 4, 46010 Valencia, SpainEndocrinology and Nutrition Department Clinic Hospital and Department of MedicineUniversity of Valencia, Valencia, SpainCIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM)Valencia, SpainDepartment of FarmacologyUniversity of Valencia, Valencia, SpainCentro de Investigación Príncipe FelipeValencia, Spain
| | - Sergio Martínez-Hervás
- Institute of Health Research-INCLIVAAvenida Menéndez Pelayo, 4, 46010 Valencia, SpainEndocrinology and Nutrition Department Clinic Hospital and Department of MedicineUniversity of Valencia, Valencia, SpainCIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM)Valencia, SpainDepartment of FarmacologyUniversity of Valencia, Valencia, SpainCentro de Investigación Príncipe FelipeValencia, Spain Institute of Health Research-INCLIVAAvenida Menéndez Pelayo, 4, 46010 Valencia, SpainEndocrinology and Nutrition Department Clinic Hospital and Department of MedicineUniversity of Valencia, Valencia, SpainCIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM)Valencia, SpainDepartment of FarmacologyUniversity of Valencia, Valencia, SpainCentro de Investigación Príncipe FelipeValencia, Spain Institute of Health Research-INCLIVAAvenida Menéndez Pelayo, 4, 46010 Valencia, SpainEndocrinology and Nutrition Department Clinic Hospital and Department of MedicineUniversity of Valencia, Valencia, SpainCIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM)Valencia, SpainDepartment of FarmacologyUniversity of Valencia, Valencia, SpainCentro de Investigación Príncipe FelipeValencia, Spain
| | - Laura Piqueras
- Institute of Health Research-INCLIVAAvenida Menéndez Pelayo, 4, 46010 Valencia, SpainEndocrinology and Nutrition Department Clinic Hospital and Department of MedicineUniversity of Valencia, Valencia, SpainCIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM)Valencia, SpainDepartment of FarmacologyUniversity of Valencia, Valencia, SpainCentro de Investigación Príncipe FelipeValencia, Spain
| | - María Jesús Sanz
- Institute of Health Research-INCLIVAAvenida Menéndez Pelayo, 4, 46010 Valencia, SpainEndocrinology and Nutrition Department Clinic Hospital and Department of MedicineUniversity of Valencia, Valencia, SpainCIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM)Valencia, SpainDepartment of FarmacologyUniversity of Valencia, Valencia, SpainCentro de Investigación Príncipe FelipeValencia, Spain Institute of Health Research-INCLIVAAvenida Menéndez Pelayo, 4, 46010 Valencia, SpainEndocrinology and Nutrition Department Clinic Hospital and Department of MedicineUniversity of Valencia, Valencia, SpainCIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM)Valencia, SpainDepartment of FarmacologyUniversity of Valencia, Valencia, SpainCentro de Investigación Príncipe FelipeValencia, Spain
| | - Deborah Jane Burks
- Institute of Health Research-INCLIVAAvenida Menéndez Pelayo, 4, 46010 Valencia, SpainEndocrinology and Nutrition Department Clinic Hospital and Department of MedicineUniversity of Valencia, Valencia, SpainCIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM)Valencia, SpainDepartment of FarmacologyUniversity of Valencia, Valencia, SpainCentro de Investigación Príncipe FelipeValencia, Spain Institute of Health Research-INCLIVAAvenida Menéndez Pelayo, 4, 46010 Valencia, SpainEndocrinology and Nutrition Department Clinic Hospital and Department of MedicineUniversity of Valencia, Valencia, SpainCIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM)Valencia, SpainDepartment of FarmacologyUniversity of Valencia, Valencia, SpainCentro de Investigación Príncipe FelipeValencia, Spain
| | - Herminia González-Navarro
- Institute of Health Research-INCLIVAAvenida Menéndez Pelayo, 4, 46010 Valencia, SpainEndocrinology and Nutrition Department Clinic Hospital and Department of MedicineUniversity of Valencia, Valencia, SpainCIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM)Valencia, SpainDepartment of FarmacologyUniversity of Valencia, Valencia, SpainCentro de Investigación Príncipe FelipeValencia, Spain Institute of Health Research-INCLIVAAvenida Menéndez Pelayo, 4, 46010 Valencia, SpainEndocrinology and Nutrition Department Clinic Hospital and Department of MedicineUniversity of Valencia, Valencia, SpainCIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM)Valencia, SpainDepartment of FarmacologyUniversity of Valencia, Valencia, SpainCentro de Investigación Príncipe FelipeValencia, Spain
| |
Collapse
|
188
|
Saponaro C, Gaggini M, Carli F, Gastaldelli A. The Subtle Balance between Lipolysis and Lipogenesis: A Critical Point in Metabolic Homeostasis. Nutrients 2015; 7:9453-74. [PMID: 26580649 PMCID: PMC4663603 DOI: 10.3390/nu7115475] [Citation(s) in RCA: 363] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/19/2015] [Accepted: 10/29/2015] [Indexed: 12/17/2022] Open
Abstract
Excessive accumulation of lipids can lead to lipotoxicity, cell dysfunction and alteration in metabolic pathways, both in adipose tissue and peripheral organs, like liver, heart, pancreas and muscle. This is now a recognized risk factor for the development of metabolic disorders, such as obesity, diabetes, fatty liver disease (NAFLD), cardiovascular diseases (CVD) and hepatocellular carcinoma (HCC). The causes for lipotoxicity are not only a high fat diet but also excessive lipolysis, adipogenesis and adipose tissue insulin resistance. The aims of this review are to investigate the subtle balances that underlie lipolytic, lipogenic and oxidative pathways, to evaluate critical points and the complexities of these processes and to better understand which are the metabolic derangements resulting from their imbalance, such as type 2 diabetes and non alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Chiara Saponaro
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, via Moruzzi, 1 56124 Pisa, Italy.
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, 53100 Siena, Italy.
| | - Melania Gaggini
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, via Moruzzi, 1 56124 Pisa, Italy.
- Dipartimento di Patologia Chirurgica, Molecolare Medica e di Area Critica, Università di Pisa, 56126 Pisa, Italy.
| | - Fabrizia Carli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, via Moruzzi, 1 56124 Pisa, Italy.
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, via Moruzzi, 1 56124 Pisa, Italy.
| |
Collapse
|
189
|
Abstract
PURPOSE OF REVIEW Metabolic diseases, such as type 2 diabetes, cardiac dysfunction, hypertension, and hepatic steatosis, share one critical causative factor: abnormal lipid partitioning, that redistribution of triglycerides from adipocytes to nonadipose peripheral tissues. Lipid overload of these tissues causes a number of pathological effects collectively known as lipotoxicity. If we find the way to correct lipid partitioning, we will restrain metabolic diseases, improve life quality and life expectancy and radically reduce healthcare costs. RECENT FINDINGS Lipid partitioning in the body is maintained by tightly regulated and balanced rates of de novo lipogenesis, lipolysis, adipogenesis, and mitochondrial oxidation primarily in fat and liver. Recent studies highlighted in this review have established mTOR as a central regulator of lipid storage and metabolism. SUMMARY Increased activity of mTOR in obesity may compensate for the negative consequences of overnutrition, whereas dysregulation of mTOR may lead to abnormal lipid partitioning and metabolic disease.
Collapse
Affiliation(s)
- Partha Chakrabarti
- aDivision of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India bDepartment of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | | |
Collapse
|
190
|
Liu HW, Chan YC, Wang MF, Wei CC, Chang SJ. Dietary (-)-Epigallocatechin-3-gallate Supplementation Counteracts Aging-Associated Skeletal Muscle Insulin Resistance and Fatty Liver in Senescence-Accelerated Mouse. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:8407-8417. [PMID: 26152236 DOI: 10.1021/acs.jafc.5b02501] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Aging is accompanied by pathophysiological changes including insulin resistance and fatty liver. Dietary supplementation with (-)-epigallocatechin-3-gallate (EGCG) improves insulin sensitivity and attenuates fatty liver disease. We hypothesized that EGCG could effectively modulate aging-associated changes in glucose and lipid metabolism in senescence-accelerated mice (SAM) prone 8 (SAMP8). Higher levels of glucose, insulin, and free fatty acid, inhibited Akt activity, and decreased glucose transporter 4 (GLUT4) expression were observed in SAMP8 mice compared to the normal aging group, SAM resistant 1 mice. EGCG supplementation for 12 weeks successfully decreased blood glucose and insulin levels via restoring Akt activity and GLUT4 expression and stimulating AMPKα activation in skeletal muscle. EGCG up-regulated genes involved in mitochondrial biogenesis and subsequently restored mitochondrial DNA copy number in skeletal muscle of SAMP8 mice. Decreased adipose triglyceride lipase and increased sterol regulatory element binding proteins-1c (SREBP-1c) and carbohydrate responsive element binding protein at mRNA levels were observed in SAMP8 mice in accordance with hepatocellular ballooning and excess lipid accumulation. The pevention of hepatic lipid accumulation by EGCG was mainly attributed to down-regulation of mTOR and SREBP-1c-mediated lipid biosynthesis via suppression of the positive regulator, Akt, and activation of the negative regulator, AMPKα, in the liver. EGCG beneficially modulates glucose and lipid homeostasis in skeletal muscle and liver, leading to alleviation of aging-associated metabolic disorders.
Collapse
Affiliation(s)
- Hung-Wen Liu
- Department of Life Sciences, National Cheng Kung University , Tainan, Taiwan
| | - Yin-Ching Chan
- Department of Food and Nutrition, Providence University , Taichung, Taiwan
| | - Ming-Fu Wang
- Department of Food and Nutrition, Providence University , Taichung, Taiwan
| | - Chu-Chun Wei
- Department of Life Sciences, National Cheng Kung University , Tainan, Taiwan
| | - Sue-Joan Chang
- Department of Life Sciences, National Cheng Kung University , Tainan, Taiwan
| |
Collapse
|
191
|
Mashek DG, Khan SA, Sathyanarayan A, Ploeger JM, Franklin MP. Hepatic lipid droplet biology: Getting to the root of fatty liver. Hepatology 2015; 62:964-7. [PMID: 25854913 PMCID: PMC4549163 DOI: 10.1002/hep.27839] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/04/2015] [Indexed: 12/22/2022]
Abstract
Hepatic steatosis is defined by the accumulation of lipid droplets (LDs). Once thought to be only inert energy storage depots, LDs are increasingly recognized as organelles that have important functions in hepatocytes beyond lipid storage. The lipid and protein composition of LDs is highly dynamic and influences their intrinsic metabolism and signaling properties, which ultimately links them to the changes in hepatic function. This concise review highlights recent discoveries in LD biology and unique aspects of hepatic LDs and their role in liver disease.
Collapse
Affiliation(s)
- Douglas G Mashek
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| | - Salmaan A Khan
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| | | | - Jonathan M Ploeger
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| | - Mallory P Franklin
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| |
Collapse
|
192
|
Bolsoni-Lopes A, Alonso-Vale MIC. Lipolysis and lipases in white adipose tissue - An update. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2015; 59:335-342. [PMID: 26331321 DOI: 10.1590/2359-3997000000067] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/22/2015] [Indexed: 11/22/2022]
Abstract
Lipolysis is defined as the sequential hydrolysis of triacylglycerol (TAG) stored in cell lipid droplets. For many years, it was believed that hormone-sensitive lipase (HSL) and monoacylglycerol lipase (MGL) were the main enzymes catalyzing lipolysis in the white adipose tissue. Since the discovery of adipose triglyceride lipase (ATGL) in 2004, many studies were performed to investigate and characterize the actions of this lipase, as well as of other proteins and possible regulatory mechanisms involved, which reformulated the concept of lipolysis. Novel findings from these studies include the identification of lipolytic products as signaling molecules regulating important metabolic processes in many non-adipose tissues, unveiling a previously underestimated aspect of lipolysis. Thus, we present here an updated review of concepts and regulation of white adipocyte lipolysis with a special emphasis in its role in metabolism homeostasis and as a source of important signaling molecules.
Collapse
Affiliation(s)
- Andressa Bolsoni-Lopes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, BR
| | - Maria Isabel C Alonso-Vale
- Departamento de Ciências Biológicas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, SP, BR
| |
Collapse
|
193
|
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
|
194
|
Matsuo T, Ushiroda Y. Fatty liver formation in fulminant type 1 diabetes. Endocrinol Diabetes Metab Case Rep 2015; 2016:15-0121. [PMID: 30367750 PMCID: PMC6356111 DOI: 10.1530/edm-15-0121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/09/2016] [Indexed: 12/02/2022] Open
Abstract
Summary A 32-year-old woman presented with 3days of epigastric pain and was admitted to our hospital (day 3 of disease). We diagnosed acute pancreatitis based on epigastric abdominal pain, hyperamylasemia, and an inflammatory reaction of withdrawn blood, pancreatic enlargement, and so on. Her condition improved with treatment; however, on day 8, she had decreased level of consciousness. Laboratory results led to a diagnosis of fulminant type 1 diabetes mellitus (FT1DM) with concomitant diabetic ketoacidosis. Insulin therapy improved her blood glucose levels as well as her symptoms. Fatty liver with liver dysfunction was observed on day 14, which improved by day 24. Blood levels of free fatty acids (FFAs) increased rapidly from 440μEq/L (normal range: 140–850μEq/L) on day 4 to 2097μEq/L on days 7–8 (onset of FT1DM) and subsequently decreased to 246μEq/L at the onset of fatty liver. The rapid decrease in insulin at the onset of FT1DM likely freed fatty acids derived from triglycerides in peripheral adipocytes into the bloodstream. Insulin therapy rapidly transferred FFAs from the periphery to the liver. In addition, insulin promotes the de novo synthesis of triglycerides in the liver, using newly acquired FFAs as substrates. At the same time, inhibitory effects of insulin on VLDL secretion outside of the liver promote the accumulation of triglycerides in the liver, leading to fatty liver. We describe the process by which liver dysfunction and severe fatty liver occurs after the onset of FT1DM, from the perspective of disturbed fatty acid metabolism. Learning Points FT1DM is rare but should be considered in patients with pancreatitis and a decreased level of consciousness. Fatty liver should be considered in patients with FT1DM when liver dysfunction is observed. Insulin is involved in mechanisms that promote fatty liver formation. Pathophysiological changes in fatty acid metabolism may provide clues on lipid metabolism in the early phases of FT1DM.
Collapse
Affiliation(s)
- Takashi Matsuo
- Internal Medicine, Nobeoka city Medical Association Hospital, Nobeoka, Japan
| | - Yoshihiko Ushiroda
- Internal Medicine, Nobeoka city Medical Association Hospital, Nobeoka, Japan
| |
Collapse
|
195
|
Tanaka N, Takahashi S, Zhang Y, Krausz KW, Smith PB, Patterson AD, Gonzalez FJ. Role of fibroblast growth factor 21 in the early stage of NASH induced by methionine- and choline-deficient diet. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1852:1242-1252. [PMID: 25736301 PMCID: PMC4433820 DOI: 10.1016/j.bbadis.2015.02.012] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 02/18/2015] [Accepted: 02/24/2015] [Indexed: 02/06/2023]
Abstract
Fibroblast growth factor 21 (FGF21) is a modulator of energy homeostasis and is increased in human nonalcoholic liver disease (NAFLD) and after feeding of methionine- and choline-deficient diet (MCD), a conventional inducer of murine nonalcoholic steatohepatitis (NASH). However, the significance of FGF21 induction in the occurrence of MCD-induced NASH remains undetermined. C57BL/6J Fgf21-null and wild-type mice were treated with MCD for 1 week. Hepatic Fgf21 mRNA was increased early after commencing MCD treatment independent of peroxisome proliferator-activated receptor (PPAR) α and farnesoid X receptor. While no significant differences in white adipose lipolysis were seen in both genotypes, hepatic triglyceride (TG) contents were increased in Fgf21-null mice, likely due to the up-regulation of genes encoding CD36 and phosphatidic acid phosphatase 2a/2c, involved in fatty acid (FA) uptake and diacylglycerol synthesis, respectively, and suppression of increased mRNAs encoding carnitine palmitoyl-CoA transferase 1α, PPARγ coactivator 1α, and adipose TG lipase, which are associated with lipid clearance in the liver. The MCD-treated Fgf21-null mice showed increased hepatic endoplasmic reticulum (ER) stress. Exposure of primary hepatocytes to palmitic acid elevated the mRNA levels encoding DNA damage-inducible transcript 3, an indicator of ER stress, and FGF21 in a PPARα-independent manner, suggesting that lipid-induced ER stress can enhance hepatic FGF21 expression. Collectively, FGF21 is elevated in the early stage of MCD-induced NASH likely to minimize hepatic lipid accumulation and ensuing ER stress. These results provide a possible mechanism on how FGF21 is increased in NAFLD/NASH.
Collapse
Affiliation(s)
- Naoki Tanaka
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States; Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Shogo Takahashi
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yuan Zhang
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Philip B Smith
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, United States
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, United States
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
196
|
Schlager S, Goeritzer M, Jandl K, Frei R, Vujic N, Kolb D, Strohmaier H, Dorow J, Eichmann TO, Rosenberger A, Wölfler A, Lass A, Kershaw EE, Ceglarek U, Dichlberger A, Heinemann A, Kratky D. Adipose triglyceride lipase acts on neutrophil lipid droplets to regulate substrate availability for lipid mediator synthesis. J Leukoc Biol 2015; 98:837-50. [PMID: 26109679 PMCID: PMC4594763 DOI: 10.1189/jlb.3a0515-206r] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/09/2015] [Indexed: 01/18/2023] Open
Abstract
Lipid mediator release depends on the hydrolysis of triglyceride-rich lipid droplets mediated by ATGL, a potent regulator of inflammatory diseases. In humans, mutations in ATGL lead to TG accumulation in LDs of most tissues and cells, including peripheral blood leukocytes. This pathologic condition is called Jordans’ anomaly, in which functional consequences have not been investigated. In the present study, we tested the hypothesis that ATGL plays a role in leukocyte LD metabolism and immune cell function. Similar to humans with loss-of-function mutations in ATGL, we found that global and myeloid-specific Atgl−/− mice exhibit Jordans’ anomaly with increased abundance of intracellular TG-rich LDs in neutrophil granulocytes. In a model of inflammatory peritonitis, lipid accumulation was also observed in monocytes and macrophages but not in eosinophils or lymphocytes. Neutrophils from Atgl−/− mice showed enhanced immune responses in vitro, which were more prominent in cells from global compared with myeloid-specific Atgl−/− mice. Mechanistically, ATGL−/− as well as pharmacological inhibition of ATGL led to an impaired release of lipid mediators from neutrophils. These findings demonstrate that the release of lipid mediators is dependent on the liberation of precursor molecules from the TG-rich pool of LDs by ATGL. Our data provide mechanistic insights into Jordans’ anomaly in neutrophils and suggest that ATGL is a potent regulator of immune cell function and inflammatory diseases.
Collapse
Affiliation(s)
- Stefanie Schlager
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Madeleine Goeritzer
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Katharina Jandl
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Robert Frei
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Nemanja Vujic
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Dagmar Kolb
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Heimo Strohmaier
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Juliane Dorow
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Thomas O Eichmann
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Angelika Rosenberger
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Albert Wölfler
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Achim Lass
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Erin E Kershaw
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Uta Ceglarek
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Andrea Dichlberger
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Akos Heinemann
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Dagmar Kratky
- Institutes of *Molecular Biology and Biochemistry and Experimental and Clinical Pharmacology, Center for Medical Research, and Division of Hematology, Medical University of Graz, Graz, Austria; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany; Institute of Molecular Biosciences, University of Graz, Graz, Austria; **Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; and Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
197
|
Li C, Li L, Lian J, Watts R, Nelson R, Goodwin B, Lehner R. Roles of Acyl-CoA:Diacylglycerol Acyltransferases 1 and 2 in Triacylglycerol Synthesis and Secretion in Primary Hepatocytes. Arterioscler Thromb Vasc Biol 2015; 35:1080-1091. [PMID: 25792450 DOI: 10.1161/atvbaha.114.304584] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 03/04/2015] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Very low-density lipoprotein assembly and secretion are regulated by the availability of triacylglycerol. Although compelling evidence indicates that the majority of triacylglycerol in very low-density lipoprotein is derived from re-esterification of lipolytic products released by endoplasmic reticulum-associated lipases, little is known about roles of acyl-CoA:diacylglycerol acyltransferases (DGATs) in this process. We aimed to investigate the contribution of DGAT1 and DGAT2 in lipid metabolism and lipoprotein secretion in primary mouse and human hepatocytes. APPROACH AND RESULTS We used highly selective small-molecule inhibitors of DGAT1 and DGAT2, and we tracked storage and secretion of lipids synthesized de novo from [(3)H]acetic acid and from exogenously supplied [(3)H]oleic acid. Inactivation of individual DGAT activity did not affect incorporation of either radiolabeled precursor into intracellular triacylglycerol, whereas combined inactivation of both DGATs severely attenuated triacylglycerol synthesis. However, inhibition of DGAT2 augmented fatty acid oxidation, whereas inhibition of DGAT1 increased triacylglycerol secretion, suggesting preferential channeling of separate DGAT-derived triacylglycerol pools to distinct metabolic pathways. Inactivation of DGAT2 impaired cytosolic lipid droplet expansion, whereas DGAT1 inactivation promoted large lipid droplet formation. Moreover, inactivation of DGAT2 attenuated expression of lipogenic genes. Finally, triacylglycerol secretion was significantly reduced on DGAT2 inhibition without altering extracellular apolipoprotein B levels. CONCLUSIONS Our data suggest that DGAT1 and DGAT2 can compensate for each other to synthesize triacylglycerol, but triacylglycerol synthesized by DGAT1 is preferentially channeled to oxidation, whereas DGAT2 synthesizes triacylglycerol destined for very low-density lipoprotein assembly.
Collapse
Affiliation(s)
- Chen Li
- From the Group on Molecular and Cell Biology of Lipids (C.L., L.L., J.L., R.W., R.N., R.L.), Department of Cell Biology (C.L., R.L.), Department of Pediatrics (L.L., J.L., R.W., R.N., R.L.), Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; and Pfizer Global Research and Development, Cardiovascular and Metabolic Diseases Research Unit, Cambridge, MA (B.G.)
| | - Lena Li
- From the Group on Molecular and Cell Biology of Lipids (C.L., L.L., J.L., R.W., R.N., R.L.), Department of Cell Biology (C.L., R.L.), Department of Pediatrics (L.L., J.L., R.W., R.N., R.L.), Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; and Pfizer Global Research and Development, Cardiovascular and Metabolic Diseases Research Unit, Cambridge, MA (B.G.)
| | - Jihong Lian
- From the Group on Molecular and Cell Biology of Lipids (C.L., L.L., J.L., R.W., R.N., R.L.), Department of Cell Biology (C.L., R.L.), Department of Pediatrics (L.L., J.L., R.W., R.N., R.L.), Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; and Pfizer Global Research and Development, Cardiovascular and Metabolic Diseases Research Unit, Cambridge, MA (B.G.)
| | - Russell Watts
- From the Group on Molecular and Cell Biology of Lipids (C.L., L.L., J.L., R.W., R.N., R.L.), Department of Cell Biology (C.L., R.L.), Department of Pediatrics (L.L., J.L., R.W., R.N., R.L.), Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; and Pfizer Global Research and Development, Cardiovascular and Metabolic Diseases Research Unit, Cambridge, MA (B.G.)
| | - Randal Nelson
- From the Group on Molecular and Cell Biology of Lipids (C.L., L.L., J.L., R.W., R.N., R.L.), Department of Cell Biology (C.L., R.L.), Department of Pediatrics (L.L., J.L., R.W., R.N., R.L.), Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; and Pfizer Global Research and Development, Cardiovascular and Metabolic Diseases Research Unit, Cambridge, MA (B.G.)
| | - Bryan Goodwin
- From the Group on Molecular and Cell Biology of Lipids (C.L., L.L., J.L., R.W., R.N., R.L.), Department of Cell Biology (C.L., R.L.), Department of Pediatrics (L.L., J.L., R.W., R.N., R.L.), Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; and Pfizer Global Research and Development, Cardiovascular and Metabolic Diseases Research Unit, Cambridge, MA (B.G.)
| | - Richard Lehner
- From the Group on Molecular and Cell Biology of Lipids (C.L., L.L., J.L., R.W., R.N., R.L.), Department of Cell Biology (C.L., R.L.), Department of Pediatrics (L.L., J.L., R.W., R.N., R.L.), Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; and Pfizer Global Research and Development, Cardiovascular and Metabolic Diseases Research Unit, Cambridge, MA (B.G.).
| |
Collapse
|
198
|
DiStefano MT, Danai LV, Roth Flach RJ, Chawla A, Pedersen DJ, Guilherme A, Czech MP. The Lipid Droplet Protein Hypoxia-inducible Gene 2 Promotes Hepatic Triglyceride Deposition by Inhibiting Lipolysis. J Biol Chem 2015; 290:15175-84. [PMID: 25922078 DOI: 10.1074/jbc.m115.650184] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Indexed: 12/22/2022] Open
Abstract
The liver is a major site of glucose, fatty acid, and triglyceride (TG) synthesis and serves as a major regulator of whole body nutrient homeostasis. Chronic exposure of humans or rodents to high-calorie diets promotes non-alcoholic fatty liver disease, characterized by neutral lipid accumulation in lipid droplets (LD) of hepatocytes. Here we show that the LD protein hypoxia-inducible gene 2 (Hig2/Hilpda) functions to enhance lipid accumulation in hepatocytes by attenuating TG hydrolysis. Hig2 expression increased in livers of mice on a high-fat diet and during fasting, two states associated with enhanced hepatic TG content. Hig2 expressed in primary mouse hepatocytes localized to LDs and promoted LD TG deposition in the presence of oleate. Conversely, tamoxifen-inducible Hig2 deletion reduced both TG content and LD size in primary hepatocytes from mice harboring floxed alleles of Hig2 and a cre/ERT2 transgene controlled by the ubiquitin C promoter. Hepatic TG was also decreased by liver-specific deletion of Hig2 in mice with floxed Hig2 expressing cre controlled by the albumin promoter. Importantly, we demonstrate that Hig2-deficient hepatocytes exhibit increased TG lipolysis, TG turnover, and fatty acid oxidation as compared with controls. Interestingly, mice with liver-specific Hig2 deletion also display improved glucose tolerance. Taken together, these data indicate that Hig2 plays a major role in promoting lipid sequestration within LDs in mouse hepatocytes through a mechanism that impairs TG degradation.
Collapse
Affiliation(s)
- Marina T DiStefano
- From the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Laura V Danai
- From the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Rachel J Roth Flach
- From the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Anil Chawla
- From the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - David J Pedersen
- From the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Adilson Guilherme
- From the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Michael P Czech
- From the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
199
|
Waterman C, Rojas-Silva P, Tumer TB, Kuhn P, Richard AJ, Wicks S, Stephens JM, Wang Z, Mynatt R, Cefalu W, Raskin I. Isothiocyanate-rich Moringa oleifera extract reduces weight gain, insulin resistance, and hepatic gluconeogenesis in mice. Mol Nutr Food Res 2015; 59:1013-24. [PMID: 25620073 DOI: 10.1002/mnfr.201400679] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 01/02/2023]
Abstract
SCOPE Moringa oleifera (moringa) is tropical plant traditionally used as an antidiabetic food. It produces structurally unique and chemically stable moringa isothiocyanates (MICs) that were evaluated for their therapeutic use in vivo. METHODS AND RESULTS C57BL/6L mice fed very high fat diet (VHFD) supplemented with 5% moringa concentrate (MC, delivering 66 mg/kg/d of MICs) accumulated fat mass, had improved glucose tolerance and insulin signaling, and did not develop fatty liver disease compared to VHFD-fed mice. MC-fed group also had reduced plasma insulin, leptin, resistin, cholesterol, IL-1β, TNFα, and lower hepatic glucose-6-phosphatase (G6P) expression. In hepatoma cells, MC and MICs at low micromolar concentrations inhibited gluconeogenesis and G6P expression. MICs and MC effects on lipolysis in vitro and on thermogenic and lipolytic genes in adipose tissue in vivo argued these are not likely primary targets for the anti-obesity and anti-diabetic effects observed. CONCLUSION Data suggest that MICs are the main anti-obesity and anti-diabetic bioactives of MC, and that they exert their effects by inhibiting rate-limiting steps in liver gluconeogenesis resulting in direct or indirect increase in insulin signaling and sensitivity. These conclusions suggest that MC may be an effective dietary food for the prevention and treatment of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Carrie Waterman
- Department of Plant Biology and Pathology, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Patricio Rojas-Silva
- Department of Plant Biology and Pathology, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Tugba Boyunegmez Tumer
- Department of Plant Biology and Pathology, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Peter Kuhn
- Department of Plant Biology and Pathology, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | | | - Shawna Wicks
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | - Zhong Wang
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Randy Mynatt
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - William Cefalu
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Ilya Raskin
- Department of Plant Biology and Pathology, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
200
|
Taschler U, Schreiber R, Chitraju C, Grabner GF, Romauch M, Wolinski H, Haemmerle G, Breinbauer R, Zechner R, Lass A, Zimmermann R. Adipose triglyceride lipase is involved in the mobilization of triglyceride and retinoid stores of hepatic stellate cells. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:937-45. [PMID: 25732851 PMCID: PMC4408194 DOI: 10.1016/j.bbalip.2015.02.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/05/2015] [Accepted: 02/22/2015] [Indexed: 01/04/2023]
Abstract
Hepatic stellate cells (HSCs) store triglycerides (TGs) and retinyl ester (RE) in cytosolic lipid droplets. RE stores are degraded following retinoid starvation or in response to pathogenic stimuli resulting in HSC activation. At present, the major enzymes catalyzing lipid degradation in HSCs are unknown. In this study, we investigated whether adipose triglyceride lipase (ATGL) is involved in RE catabolism of HSCs. Additionally, we compared the effects of ATGL deficiency and hormone-sensitive lipase (HSL) deficiency, a known RE hydrolase (REH), on RE stores in liver and adipose tissue. We show that ATGL degrades RE even in the presence of TGs, implicating that these substrates compete for ATGL binding. REH activity was stimulated and inhibited by comparative gene identification-58 and G0/G1 switch gene-2, respectively, the physiological regulators of ATGL activity. In cultured primary murine HSCs, pharmacological inhibition of ATGL, but not HSL, increased RE accumulation. In mice globally lacking ATGL or HSL, RE contents in white adipose tissue were decreased or increased, respectively, while plasma retinol and liver RE levels remained unchanged. In conclusion, our study shows that ATGL acts as REH in HSCs promoting the degradation of RE stores in addition to its established function as TG lipase. HSL is the predominant REH in adipocytes but does not affect lipid mobilization in HSCs. ATGL possesses retinyl ester and triacylglycerol hydrolase activity. The lack of ATGL activity causes increased triacylglycerol and retinyl ester storage in hepatic stellate cells. ATGL acts as retinyl ester and triacylglycerol lipase in hepatic stellate cells.
Collapse
Affiliation(s)
- Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, Graz 8010, Austria
| | - Renate Schreiber
- Institute of Molecular Biosciences, University of Graz, Graz 8010, Austria
| | | | - Gernot F Grabner
- Institute of Molecular Biosciences, University of Graz, Graz 8010, Austria
| | - Matthias Romauch
- Institute of Molecular Biosciences, University of Graz, Graz 8010, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, Graz 8010, Austria
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, Graz 8010, Austria
| | - Rolf Breinbauer
- Institute of Organic Chemistry, Graz University of Technology, Graz 8010, Austria
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Graz 8010, Austria
| | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Graz 8010, Austria.
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz 8010, Austria.
| |
Collapse
|