1
|
Uchiyama LF, Ordonez GPM, Pham KT, Kennelly JP, López Rodríguez M, Tran L, Tontonoz P, Nguyen A. PPARɑ variant V227A reduces plasma triglycerides through enhanced lipoprotein lipolysis. J Lipid Res 2025; 66:100806. [PMID: 40245984 DOI: 10.1016/j.jlr.2025.100806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025] Open
Abstract
Human single nucleotide variants in peroxisome proliferator-activated receptor-ɑ (PPARɑ) have been associated with beneficial metabolic phenotypes, yet their specific effects on metabolic gene expression are not well defined. Here, we developed a mouse model of a human PPARɑ variant encoding a substitution of valine for alanine at position 227 (V227A) to explore the role of this variant on systemic metabolism. Substitution with this variant in mice reduced plasma triglycerides, without altering body mass or liver lipid accumulation, consistent with phenotypes observed in human cohorts. Gene expression analysis revealed that the V227A variant enhances Ppara target gene expression in mouse liver, consistent with the effects of synthetic PPARɑ agonist treatment. Notably, V227A increased hepatic expression of Lpl, the predominant enzyme responsible for circulating triglyceride hydrolysis. Further characterization revealed that heart tissue from variant mice exhibited increased Lpl expression and triglyceride hydrolysis activity, suggesting that V227A enhances cardiac triglyceride clearance. These findings validate human observational studies and clarify the physiological impact of the V227A PPARɑ variant on plasma triglycerides.
Collapse
Affiliation(s)
- Lauren F Uchiyama
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA; Department of Biological Chemistry, University of California, Los Angeles, CA
| | - Gabriel P M Ordonez
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Khoi T Pham
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - John P Kennelly
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA; Department of Biological Chemistry, University of California, Los Angeles, CA
| | - Maykel López Rodríguez
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA; Department of Biological Chemistry, University of California, Los Angeles, CA
| | - Lany Tran
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA; Department of Biological Chemistry, University of California, Los Angeles, CA
| | - Alexander Nguyen
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA.
| |
Collapse
|
2
|
Xia Z, Gong G, Huang R, Goossens T, Lemâle O, Cardoso D, Mallmann B, Zhao L, Wei J, Deng J. Butyric acid-based products, alone or in combination with hydroxy-selenomethionine, improve performance of laying hens in post-peak period by modulating their antioxidant, metabolic and immune status. Poult Sci 2025; 104:104840. [PMID: 40020406 PMCID: PMC11919429 DOI: 10.1016/j.psj.2025.104840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 03/03/2025] Open
Abstract
This study aimed to evaluate the effects of butyrate, butyric glycerides (BG) and their combination with sodium selenite (SeNa) or hydroxy-selenomethionine (OH-SeMet) on the performance and egg quality of the laying hens in post-peak period as well as the potential mechanism. A total of 900 45-week-old Hy-Line brown laying hens were randomly allocated to 5 treatment groups (n = 10 replicates/diet, 18 hens/replicate). The hens were fed the basal diet supplemented with 0.3 mg/kg selenium from SeNa (Control), Control plus 240 mg/kg butyric acid from coated butyrate (CB), Control plus 240 mg/kg butyric acid from butyric glycerides, basal diet supplemented with 0.3 mg/kg selenium from OH-SeMet plus coated butyrate (CB+OH-SeMet) or butyric glycerides (BG+OH-SeMet), respectively, for 20 weeks. Serum, liver, isthmus, uterus, and jejunum were collected at the end of the trial for biochemistry, histology, redox status, and gene expression analysis. Compared with Control, diets supplemented with BG, CB+OH-SeMet and BG+OH-SeMet increased (p < 0.05) the average egg weight (0.6-2.2 %), while only BG+OH-SeMet increased (p < 0.05) the total egg weight (7.1 %) and egg-laying rate (4.6 %) and decreased (p < 0.05) the feed/egg ratio (5.0 %) throughout the whole experiment. Furthermore, BG+OH-SeMet reduced (p < 0.05) the content of IL-6 and alanine aminotransferase (15.4-32.5 %), while elevated (p < 0.05) the content of IgA, IgY, IgM and total protein (18.7-26.8 %) in the serum in comparison to the Control. Notably, dietary supplementation of BG+OH-SeMet performed more effective antioxidant capacity in decreasing (p < 0.05) malondialdehyde (16.4-27.9 %) content and increasing (p < 0.05) the activity of total antioxidant capacity and glutathione peroxidase (17.6-36.3 %) in various tissues. Further experiment revealed that dietary BG+OH-SeMet regulated the lipid metabolism by increasing (p < 0.05) the expression of Carnitine palmitoyltransferase 1A (CPT1A) and Lipoprotein lipase (LPL) in liver. In conclusion, diets supplemented with BG and OH-SeMet could improve the laying performance via the enhancement of antioxidant capacity and regulation of lipid metabolism.
Collapse
Affiliation(s)
- Zhiyuan Xia
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Guoxin Gong
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Ronghui Huang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | | | - Olga Lemâle
- Adisseo NL, 4941 SB Raamsdonksveer, The Netherlands
| | | | | | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jintao Wei
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan 430064, China.
| | - Jiang Deng
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan 430064, China.
| |
Collapse
|
3
|
Moolsup F, Suttithumsatid W, Woonnoi W, Chonpathompikunlert P, Tanasawet S, Sukketsiri W. Passion Fruit Seed Extract Attenuates Hepatic Steatosis in Oleic Acid-Treated HepG2 Cells through Modulation of ERK1/2 and Akt Signaling Pathways. Cell Biochem Biophys 2025:10.1007/s12013-025-01706-5. [PMID: 40025286 DOI: 10.1007/s12013-025-01706-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2025] [Indexed: 03/04/2025]
Abstract
Hepatic steatosis, commonly referred to as fatty liver disease, is defined by the abnormal buildup of fat within liver cells. Currently, primary treatments mainly focus on lifestyle changes, underscoring a lack of direct pharmacological options. Passion fruit seed extract (PFSE) has been reported to decrease hepatosteatosis; however, the mechanism underlying this effect has not been clarified. Therefore, the objective of this research was to investigate the effects and mechanisms of action of PFSE against oleic acid (OA)-induced hepatosteatosis in HepG2 cells. OA-induced HepG2 cells were analyzed by using various cell-based experiments, including assessments of cytotoxicity, reactive oxygen species (ROS) production, apoptosis, and protein and gene expression. LC-MS-MS analysis showed that PFSE contains a variety of phytochemical compounds such as alkaloids, flavonoids, stilbenoids, coumarins, terpenoids, lipids, and fatty acid derivatives, which have the potential to exhibit various pharmacological activities. In this study, PFSE demonstrated antioxidant, anti-inflammatory, and lipid metabolism-regulating activities. It also influenced key genes related to lipid metabolism, including SREBP-1c, ACC, FASN, PPARα, CPT-1A, LPL, SCD1, and LDLR. The positive effects of PFSE on OA-induced hepatic steatosis in HepG2 cells were modulated through the Akt and ERK signaling pathways, suggesting that PFSE may offer a comprehensive approach to managing hepatic steatosis.
Collapse
Affiliation(s)
- Furoida Moolsup
- Laboratory Animal Service Center, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
| | - Wiwit Suttithumsatid
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla, Thailand
- Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Thailand
| | - Wanwipha Woonnoi
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Pennapa Chonpathompikunlert
- Research and Development Group for Bio-Industries, Thailand Institute of Scientific and Technological Research (TISTR), Pathumthani, Thailand
| | - Supita Tanasawet
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Wanida Sukketsiri
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand.
| |
Collapse
|
4
|
Shen J, Liang W, Zhao R, Chen Y, Liu Y, Cheng W, Chai T, Zhang Y, Chen S, Liu J, Chen X, Deng Y, Zhang Z, Huang Y, Yang H, Pang L, Qiu Q, Deng H, Pan S, Wang L, Ye J, Luo W, Jiang X, Huang X, Li W, Leung EL, Zhang L, Huang L, Yang Z, Chen R, Mei J, Yue Z, Wei H, Karsten K, Han L, Fang X. Cross-tissue multi-omics analyses reveal the gut microbiota's absence impacts organ morphology, immune homeostasis, bile acid and lipid metabolism. IMETA 2025; 4:e272. [PMID: 40027481 PMCID: PMC11865341 DOI: 10.1002/imt2.272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 03/05/2025]
Abstract
The gut microbiota influences host immunity and metabolism, and changes in its composition and function have been implicated in several non-communicable diseases. Here, comparing germ-free (GF) and specific pathogen-free (SPF) mice using spatial transcriptomics, single-cell RNA sequencing, and targeted bile acid metabolomics across multiple organs, we systematically assessed how the gut microbiota's absence affected organ morphology, immune homeostasis, bile acid, and lipid metabolism. Through integrated analysis, we detect marked aberration in B, myeloid, and T/natural killer cells, altered mucosal zonation and nutrient uptake, and significant shifts in bile acid profiles in feces, liver, and circulation, with the alternate synthesis pathway predominant in GF mice and pronounced changes in bile acid enterohepatic circulation. Particularly, autophagy-driven lipid droplet breakdown in ileum epithelium and the liver's zinc finger and BTB domain-containing protein (ZBTB20)-Lipoprotein lipase (LPL) (ZBTB20-LPL) axis are key to plasma lipid homeostasis in GF mice. Our results unveil the complexity of microbiota-host interactions in the crosstalk between commensal gut bacteria and the host.
Collapse
Affiliation(s)
- Juan Shen
- BGI ResearchShenzhenChina
- Qingdao‐Europe Advanced Institute for Life SciencesBGI ResearchQingdaoChina
| | | | | | - Yang Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine SyndromeThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Yanmin Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine SyndromeThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Wei Cheng
- College of Animal Sciences and TechnologyHuazhong Agricultural UniversityWuhanChina
| | | | | | | | | | | | - Yusheng Deng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine SyndromeThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | | | | | | | | | - Qinwei Qiu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine SyndromeThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | | | | | | | | | - Wen Luo
- Kangmeihuada (KMHD) GeneTech Co., Ltd.ShenzhenChina
- Zhuhai UM Science & Technology Research Institute‐Kangmeihuada (KMHD) joint labZhuhaiChina
| | - Xuanting Jiang
- Kangmeihuada (KMHD) GeneTech Co., Ltd.ShenzhenChina
- Zhuhai UM Science & Technology Research Institute‐Kangmeihuada (KMHD) joint labZhuhaiChina
| | | | | | - Elaine Lai‐Han Leung
- Zhuhai UM Science & Technology Research Institute‐Kangmeihuada (KMHD) joint labZhuhaiChina
- Cancer Center, Faculty of Health SciencesUniversity of MacauMacau (SAR)China
- MOE Frontiers Science Center for Precision OncologyUniversity of MacauMacau (SAR)China
| | - Lu Zhang
- Department of Computer ScienceHong Kong Baptist UniversityHong KongChina
| | - Li Huang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine SyndromeThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Zhimin Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine SyndromeThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | | | - Junpu Mei
- BGI ResearchShenzhenChina
- BGI ResearchSanyaChina
| | | | - Hong Wei
- College of Animal Sciences and TechnologyHuazhong Agricultural UniversityWuhanChina
- Yu‐Yue Pathology Scientific Research CenterChongqingChina
| | - Kristiansen Karsten
- BGI ResearchShenzhenChina
- Laboratory of Genomics and Molecular Biomedicine, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Lijuan Han
- Kangmeihuada (KMHD) GeneTech Co., Ltd.ShenzhenChina
- Zhuhai UM Science & Technology Research Institute‐Kangmeihuada (KMHD) joint labZhuhaiChina
- Kangmei Pharmaceutical Co., Ltd.JieyangChina
| | - Xiaodong Fang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine SyndromeThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
- BGI ResearchSanyaChina
| |
Collapse
|
5
|
Kineman RD, Del Rio-Moreno M, Waxman DJ. Liver-specific actions of GH and IGF1 that protect against MASLD. Nat Rev Endocrinol 2025; 21:105-117. [PMID: 39322791 DOI: 10.1038/s41574-024-01037-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD; also known as nonalcoholic fatty liver disease) is a chronic condition associated with metabolic syndrome, a group of conditions that includes obesity, insulin resistance, hyperlipidaemia and cardiovascular disease. Primary growth hormone (GH) deficiency is associated with MASLD, and the decline in circulating levels of GH with weight gain might contribute to the development of MASLD. Raising endogenous GH secretion or administering GH replacement therapy in the context of MASLD enhances insulin-like growth factor 1 (IGF1) production and reduces steatosis and the severity of liver injury. GH and IGF1 indirectly control MASLD progression by regulating systemic metabolic function. Evidence supports the proposal that GH and IGF1 also have a direct role in regulating liver metabolism and health. This Review focuses on how GH acts on the hepatocyte in a sex-dependent manner to limit lipid accumulation, reduce stress, and promote survival and regeneration. In addition, we discuss how GH and IGF1 might regulate non-parenchymal cells of the liver to control inflammation and fibrosis, which have a major effect on hepatocyte survival and regeneration. Development of a better understanding of how GH and IGF1 coordinate the functions of specific, individual liver cell types might provide insight into the aetiology of MASLD initiation and progression and suggest novel approaches for the treatment of MASLD.
Collapse
Affiliation(s)
- Rhonda D Kineman
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA.
- Jesse Brown VA Medical Center, Research and Development Division, Chicago, IL, USA.
| | - Mercedes Del Rio-Moreno
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown VA Medical Center, Research and Development Division, Chicago, IL, USA
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
6
|
Bradić I, Kuentzel KB, Pirchheim A, Rainer S, Schwarz B, Trauner M, Larsen MR, Vujić N, Kratky D. From LAL-D to MASLD: Insights into the role of LAL and Kupffer cells in liver inflammation and lipid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159575. [PMID: 39486573 DOI: 10.1016/j.bbalip.2024.159575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent liver pathology worldwide, closely associated with obesity and metabolic disorders. Increasing evidence suggests that macrophages play a crucial role in the development of MASLD. Several human studies have shown an inverse correlation between circulating lysosomal acid lipase (LAL) activity and MASLD. LAL is the sole enzyme known to degrade cholesteryl esters (CE) and triacylglycerols in lysosomes. Consequently, these substrates accumulate when their enzymatic degradation is impaired due to LAL deficiency (LALD). This study aimed to investigate the role of hepatic LAL activity and liver-resident macrophages (i.e., Kupffer cells (KC)) in MASLD. To this end, we analyzed lipid metabolism in hepatocyte-specific (hep)Lal-/- mice and depleted KC with clodronate treatment. When fed a high-fat/high-cholesterol diet (HF/HCD), hepLal-/- mice exhibited CE accumulation and an increased number of macrophages in the liver and significant hepatic inflammation. KC were depleted upon clodronate administration, whereas infiltrating/proliferating CD68-expressing macrophages were less affected. This led to exacerbated hepatic CE accumulation and dyslipidemia, as evidenced by increased LDL-cholesterol concentrations. Along with proteomic analysis of liver tissue, these findings indicate that hepatic LAL-D in HF/HCD-fed mice leads to macrophage infiltration into the liver and that KC depletion further exacerbates hepatic CE concentrations and dyslipidemia.
Collapse
Affiliation(s)
- Ivan Bradić
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Katharina B Kuentzel
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Anita Pirchheim
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Silvia Rainer
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Birgit Schwarz
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Nemanja Vujić
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
7
|
Lincoln K, Zhou J, Oster H, de Assis LVM. Circadian Gating of Thyroid Hormone Action in Hepatocytes. Cells 2024; 13:1038. [PMID: 38920666 PMCID: PMC11202020 DOI: 10.3390/cells13121038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Thyroid hormones, thyroxin (T4) and the biologically active triiodothyronine (T3), play important roles in liver metabolic regulation, including fatty acid biosynthesis, beta-oxidation, and cholesterol homeostasis. These functions position TH signaling as a potential target for the treatment of metabolic dysfunction-associated steatotic liver disease (MASLD). Elevated T3 levels in the circulation are associated with increased hepatic lipid turnover, which is also under the control of the circadian clock system. In this study, we developed a cell system to study the impact of hepatocyte circadian rhythms on the metabolic response to T3 treatment under control and steatotic conditions. Synchronized AML-12 circadian reporter hepatocytes were treated with T3 at different circadian phases and metabolic conditions. T3 treatment increased metabolic activity in a dose-independent fashion and had no significant effect on circadian rhythms in AML-12 cells. T3 had marked time-of-treatment-dependent effects on metabolic transcript expression. Steatosis induction altered metabolic transcript expression in AML-12 cells. In this condition, the circadian rhythm period was lengthened, and this effect was independent of T3. Under steatotic conditions, T3 had marked time-of-treatment dependent effects on metabolic transcript expression, which differed from those observed under control conditions. These findings reveal a time-of-day-dependent response of hepatocytes to T3, which is further modulated by the metabolic state. Our data suggest that time has a strong influence on liver TH action, which might be considered when treating MASLD.
Collapse
Affiliation(s)
- Karla Lincoln
- Institute of Neurobiology, Center of Brain Behavior & Metabolism, University of Lübeck, 23562 Lübeck, Germany; (K.L.); (J.Z.)
| | - Jingxuan Zhou
- Institute of Neurobiology, Center of Brain Behavior & Metabolism, University of Lübeck, 23562 Lübeck, Germany; (K.L.); (J.Z.)
| | - Henrik Oster
- Institute of Neurobiology, Center of Brain Behavior & Metabolism, University of Lübeck, 23562 Lübeck, Germany; (K.L.); (J.Z.)
- University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - Leonardo Vinicius Monteiro de Assis
- Institute of Neurobiology, Center of Brain Behavior & Metabolism, University of Lübeck, 23562 Lübeck, Germany; (K.L.); (J.Z.)
- University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| |
Collapse
|
8
|
Zhang J, Xu X, Kuang H, Xu C, Wu X. Potential health risk analysis of chlorantraniliprole in vivo. Sci Bull (Beijing) 2023; 68:2712-2716. [PMID: 37758618 DOI: 10.1016/j.scib.2023.08.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/29/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023]
Affiliation(s)
- Jia Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; International Joint Research Laboratory for Biointerface and Biodetection and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Xinxin Xu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; International Joint Research Laboratory for Biointerface and Biodetection and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
| | - Hua Kuang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; International Joint Research Laboratory for Biointerface and Biodetection and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Chuanlai Xu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; International Joint Research Laboratory for Biointerface and Biodetection and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Xiaoling Wu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; International Joint Research Laboratory for Biointerface and Biodetection and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
9
|
Hu T, Li Z, Gong C, Xiong Y, Sun S, Xing J, Li Y, Li R, Wang Y, Wang Y, Lin Y. FOS Inhibits the Differentiation of Intramuscular Adipocytes in Goats. Genes (Basel) 2023; 14:2088. [PMID: 38003034 PMCID: PMC10671551 DOI: 10.3390/genes14112088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Goat intramuscular fat (IMF) deposition is precisely regulated by many key genes as well as transcription factors. Nevertheless, the potential of the regulators of goat IMF deposition remains undefined. In this work, we reported that the transcription factor FOS is expressed at a low level at the early differentiation stage and at a high level in late differentiation. The overexpression of FOS inhibited intramuscular adipocyte lipid accumulation and significantly downregulated the expressions of PPARγ, C/EBPβ, C/EBPα, AP2, SREBP1, FASN, ACC, HSL, and ATGL. Consistently, the knockdown of FOS, facilitated by two distinct siRNAs, significantly promoted intramuscular adipocyte lipid accumulation. Moreover, our analysis revealed multiple potential binding sites for FOS on the promoters of PPARγ, C/EBPβ, and C/EBPα. The expression changes in PPARγ, C/EBPβ, and C/EBPα during intramuscular adipogenesis were opposite to that of FOS. In summary, FOS inhibits intramuscular lipogenesis in goats and potentially negatively regulates the expressions of PPARγ, C/EBPβ, and C/EBPα genes. Our research will provide valuable data for the underlying molecular mechanism of the FOS regulation network of intramuscular lipogenesis.
Collapse
Affiliation(s)
- Tingting Hu
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Zhibin Li
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Chengsi Gong
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Yan Xiong
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Shiyu Sun
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
| | - Jiani Xing
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Yanyan Li
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Ruiwen Li
- Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 611731, China;
| | - Youli Wang
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Yong Wang
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Yaqiu Lin
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| |
Collapse
|
10
|
Kweon SM, Irimia-Dominguez J, Kim G, Fueger PT, Asahina K, Lai KK, Allende DS, Lai QR, Lou CH, Tsark WM, Yang JD, Ng DS, Lee JS, Tso P, Huang W, Lai KKY. Heterozygous midnolin knockout attenuates severity of nonalcoholic fatty liver disease in mice fed a Western-style diet high in fat, cholesterol, and fructose. Am J Physiol Gastrointest Liver Physiol 2023; 325:G147-G157. [PMID: 37129245 PMCID: PMC10393367 DOI: 10.1152/ajpgi.00011.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 05/03/2023]
Abstract
Although midnolin has been studied for over 20 years, its biological roles in vivo remain largely unknown, especially due to the lack of a functional animal model. Indeed, given our recent discovery that the knockdown of midnolin suppresses liver cancer cell tumorigenicity and that this antitumorigenic effect is associated with modulation of lipid metabolism, we hypothesized that knockout of midnolin in vivo could potentially protect from nonalcoholic fatty liver disease (NAFLD) which has become the most common cause of chronic liver disease in the Western world. Accordingly, in the present study, we have developed and now report on the first functional global midnolin knockout mouse model. Although the overwhelming majority of global homozygous midnolin knockout mice demonstrated embryonic lethality, heterozygous knockout mice were observed to be similar to wild-type mice in their viability and were used to determine the effect of reduced midnolin expression on NAFLD. We found that global heterozygous midnolin knockout attenuated the severity of NAFLD in mice fed a Western-style diet, high in fat, cholesterol, and fructose, and this attenuation in disease was associated with significantly reduced levels of large lipid droplets, hepatic free cholesterol, and serum LDL, with significantly differential gene expression involved in cholesterol/lipid metabolism. Collectively, our results support a role for midnolin in regulating cholesterol/lipid metabolism in the liver. Thus, midnolin may represent a novel therapeutic target for NAFLD. Finally, our observation that midnolin was essential for survival underscores the broad importance of this gene beyond its role in liver biology.NEW & NOTEWORTHY We have developed and now report on the first functional global midnolin knockout mouse model. We found that global heterozygous midnolin knockout attenuated the severity of nonalcoholic fatty liver disease (NAFLD) in mice fed a Western-style diet, high in fat, cholesterol, and fructose, and this attenuation in disease was associated with significantly reduced levels of large lipid droplets, hepatic free cholesterol, and serum LDL, with significantly differential gene expression involved in cholesterol/lipid metabolism.
Collapse
Affiliation(s)
- Soo-Mi Kweon
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, California, United States
| | - Jose Irimia-Dominguez
- Department of Molecular and Cellular Endocrinology and Comprehensive Metabolic Phenotyping Core, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, California, United States
| | - Gayeoun Kim
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, California, United States
| | - Patrick T Fueger
- Department of Molecular and Cellular Endocrinology and Comprehensive Metabolic Phenotyping Core, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, California, United States
- City of Hope Comprehensive Cancer Center, Duarte, California, United States
| | - Kinji Asahina
- Central Research Laboratory, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Japan
| | - Keith K Lai
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio, United States
- Contra Costa Pathology Associates, Pleasant Hill, California, United States
| | - Daniela S Allende
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio, United States
| | - Quincy R Lai
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, California, United States
| | - Chih-Hong Lou
- Gene Editing and Viral Vector Core, Beckman Research Institute of City of Hope, Duarte, California, United States
| | - Walter M Tsark
- Transgenic/Knockout Mouse Program, Center for Comparative Medicine, Beckman Research Institute of City of Hope, Duarte, California, United States
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, United States
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Dominic S Ng
- Departments of Medicine, Physiology, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, California, United States
- City of Hope Comprehensive Cancer Center, Duarte, California, United States
| | - Keane K Y Lai
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, California, United States
- City of Hope Comprehensive Cancer Center, Duarte, California, United States
| |
Collapse
|
11
|
Schmalz F, Fischer J, Innes H, Buch S, Möller C, Matz-Soja M, von Schönfels W, Krämer B, Langhans B, Klüners A, Soyka M, Stickel F, Nattermann J, Strassburg CP, Berg T, Lutz P, Nischalke HD. High producer variant of lipoprotein lipase may protect from hepatocellular carcinoma in alcohol-associated cirrhosis. JHEP Rep 2023; 5:100684. [PMID: 36879887 PMCID: PMC9985032 DOI: 10.1016/j.jhepr.2023.100684] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 01/11/2023] [Accepted: 01/14/2023] [Indexed: 01/26/2023] Open
Abstract
Background & Aims Progression of alcohol-associated liver disease (ALD) is driven by genetic predisposition. The rs13702 variant in the lipoprotein lipase (LPL) gene is linked to non-alcoholic fatty liver disease. We aimed at clarifying its role in ALD. Methods Patients with alcohol-associated cirrhosis, with (n = 385) and without hepatocellular carcinoma (HCC) (n = 656), with HCC attributable to viral hepatitis C (n = 280), controls with alcohol abuse without liver damage (n = 366), and healthy controls (n = 277) were genotyped regarding the LPL rs13702 polymorphism. Furthermore, the UK Biobank cohort was analysed. LPL expression was investigated in human liver specimens and in liver cell lines. Results Frequency of the LPL rs13702 CC genotype was lower in ALD with HCC in comparison to ALD without HCC both in the initial (3.9% vs. 9.3%) and the validation cohort (4.7% vs. 9.5%; p <0.05 each) and compared with patients with viral HCC (11.4%), alcohol misuse without cirrhosis (8.7%), or healthy controls (9.0%). This protective effect (odds ratio [OR] = 0.5) was confirmed in multivariate analysis including age (OR = 1.1/year), male sex (OR = 3.0), diabetes (OR = 1.8), and carriage of the PNPLA3 I148M risk variant (OR = 2.0). In the UK Biobank cohort, the LPL rs13702 C allele was replicated as a risk factor for HCC. Liver expression of LPL mRNA was dependent on LPL rs13702 genotype and significantly higher in patients with ALD cirrhosis compared with controls and alcohol-associated HCC. Although hepatocyte cell lines showed negligible LPL protein expression, hepatic stellate cells and liver sinusoidal endothelial cells expressed LPL. Conclusions LPL is upregulated in the liver of patients with alcohol-associated cirrhosis. The LPL rs13702 high producer variant confers protection against HCC in ALD, which might help to stratify people for HCC risk. Impact and implications Hepatocellular carcinoma is a severe complication of liver cirrhosis influenced by genetic predisposition. We found that a genetic variant in the gene encoding lipoprotein lipase reduces the risk for hepatocellular carcinoma in alcohol-associated cirrhosis. This genetic variation may directly affect the liver, because, unlike in healthy adult liver, lipoprotein lipase is produced from liver cells in alcohol-associated cirrhosis.
Collapse
Key Words
- ALD, alcohol-associated liver disease
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- Alcohol-associated liver disease
- BCLC, Barcelona Clinic Liver Cancer
- BSA, bovine serum albumin
- Cirrhosis
- FCS, foetal calf serum
- FIB-4, fibrosis 4
- GADPH, glyceraldehyde 3-phosphate dehydrogenase
- GGT, gamma-glutamyl transferase
- HCC
- HCC, hepatocellular carcinoma
- HSCs, hepatic stellate cells
- HbA1c, glycated haemoglobin
- LPL
- LPL, lipoprotein lipase
- LSECs, liver sinusoidal endothelial cells
- MAF, minor allele frequency
- NAFLD, non-alcoholic fatty liver disease
- NASH, non-alcoholic steatohepatitis
- OR, odds ratio
- PNPLA3, patatin-like phospholipase domain-containing protein 3
- T2DM, type 2 diabetes mellitus
- UKB, UK Biobank
- rs13702
- rs328
Collapse
Affiliation(s)
- Franziska Schmalz
- Department of Internal Medicine I, University Hospital, University of Bonn, Germany
| | - Janett Fischer
- Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| | - Hamish Innes
- School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Stephan Buch
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
| | - Christine Möller
- Department of Internal Medicine I, University Hospital, University of Bonn, Germany
| | - Madlen Matz-Soja
- Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| | - Witigo von Schönfels
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, and Christian-Albrecht University (CAU), Kiel, Germany
| | - Benjamin Krämer
- Department of Internal Medicine I, University Hospital, University of Bonn, Germany
| | - Bettina Langhans
- Department of Internal Medicine I, University Hospital, University of Bonn, Germany
| | - Alexandra Klüners
- Department of Internal Medicine I, University Hospital, University of Bonn, Germany
| | - Michael Soyka
- Psychiatric Hospital, Ludwig Maximilians University, Munich, Germany
| | - Felix Stickel
- Department of Gastroenterology and Hepatology, University Hospital of Zürich, Switzerland
| | - Jacob Nattermann
- Department of Internal Medicine I, University Hospital, University of Bonn, Germany
| | | | - Thomas Berg
- Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| | - Philipp Lutz
- Department of Internal Medicine I, University Hospital, University of Bonn, Germany
| | | |
Collapse
|
12
|
Vos DY, Wijers M, Smit M, Huijkman N, Kloosterhuis NJ, Wolters JC, Tissink JJ, Pronk ACM, Kooijman S, Rensen PCN, Kuivenhoven JA, van de Sluis B. Cargo-Specific Role for Retriever Subunit VPS26C in Hepatocyte Lipoprotein Receptor Recycling to Control Postprandial Triglyceride-Rich Lipoproteins. Arterioscler Thromb Vasc Biol 2023; 43:e29-e45. [PMID: 36353989 DOI: 10.1161/atvbaha.122.318169] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND The copper metabolism MURR1 domains/coiled-coil domain containing 22/coiled-coil domain containing 93 (CCC) complex is required for the transport of low-density lipoprotein receptor (LDLR) and LRP1 (LDLR-related protein 1) from endosomes to the cell surface of hepatocytes. Impaired functioning of hepatocytic CCC causes hypercholesterolemia in mice, dogs, and humans. Retriever, a protein complex consisting of subunits VPS26C, VPS35L, and VPS29, is associated with CCC, but its role in endosomal lipoprotein receptor transport is unclear. We here investigated the contribution of retriever to hepatocytic lipoprotein receptor recycling and plasma lipids regulation. METHODS Using somatic CRISPR/Cas9 gene editing, we generated liver-specific VPS35L or VPS26C-deficient mice. We determined total and surface levels of LDLR and LRP1 and plasma lipids. In addition, we studied the protein levels and composition of CCC and retriever. RESULTS Hepatocyte VPS35L deficiency reduced VPS26C levels but had minimal impact on CCC composition. VPS35L deletion decreased hepatocytic surface expression of LDLR and LRP1, accompanied by a 21% increase in plasma cholesterol levels. Hepatic VPS26C ablation affected neither levels of VPS35L and CCC subunits, nor plasma lipid concentrations. However, VPS26C deficiency increased hepatic LDLR protein levels by 2-fold, probably compensating for reduced LRP1 functioning, as we showed in VPS26C-deficient hepatoma cells. Upon PCSK9 (proprotein convertase subtilisin/kexin type 9)-mediated LDLR elimination, VPS26C ablation delayed postprandial triglyceride clearance and increased plasma triglyceride levels by 26%. CONCLUSIONS Our study suggests that VPS35L is shared between retriever and CCC to facilitate LDLR and LRP1 transport from endosomes to the cell surface. Conversely, retriever subunit VPS26C selectively transports LRP1, but not LDLR, and thereby may control hepatic uptake of postprandial triglyceride-rich lipoprotein remnants.
Collapse
Affiliation(s)
- Dyonne Y Vos
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands (D.Y.V., M.W., M.S., N.H., N.J.K., J.C.W., J.AK., B.v.d.S.)
| | - Melinde Wijers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands (D.Y.V., M.W., M.S., N.H., N.J.K., J.C.W., J.AK., B.v.d.S.)
| | - Marieke Smit
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands (D.Y.V., M.W., M.S., N.H., N.J.K., J.C.W., J.AK., B.v.d.S.)
| | - Nicolette Huijkman
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands (D.Y.V., M.W., M.S., N.H., N.J.K., J.C.W., J.AK., B.v.d.S.)
| | - Niels J Kloosterhuis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands (D.Y.V., M.W., M.S., N.H., N.J.K., J.C.W., J.AK., B.v.d.S.)
| | - Justina C Wolters
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands (D.Y.V., M.W., M.S., N.H., N.J.K., J.C.W., J.AK., B.v.d.S.)
| | - Joël J Tissink
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany. Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Germany (J.J.T.).,German Center for Diabetes Research (DZD), Neuherberg, Germany (J.J.T.)
| | - Amanda C M Pronk
- Department of Medicine, Division of Endocrinology (A.C.M.P., S.K., P.C.N.R.), Leiden University Medical Center, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine (A.C.M.P., S.K., P.C.N.R.), Leiden University Medical Center, the Netherlands
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology (A.C.M.P., S.K., P.C.N.R.), Leiden University Medical Center, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine (A.C.M.P., S.K., P.C.N.R.), Leiden University Medical Center, the Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology (A.C.M.P., S.K., P.C.N.R.), Leiden University Medical Center, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine (A.C.M.P., S.K., P.C.N.R.), Leiden University Medical Center, the Netherlands
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands (D.Y.V., M.W., M.S., N.H., N.J.K., J.C.W., J.AK., B.v.d.S.)
| | - Bart van de Sluis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands (D.Y.V., M.W., M.S., N.H., N.J.K., J.C.W., J.AK., B.v.d.S.)
| |
Collapse
|
13
|
Huang X, Zhuo Y, Jiang D, Zhu Y, Fang Z, Che L, Lin Y, Xu S, Hua L, Zou Y, Huang C, Li L, Wu D, Feng B. Maternal Low-Protein Diet during Puberty and Adulthood Aggravates Lipid Metabolism of Their Offspring Fed a High-Fat Diet in Mice. Nutrients 2022; 14:nu14194057. [PMID: 36235710 PMCID: PMC9570549 DOI: 10.3390/nu14194057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
A maternal low-protein (LP) diet during gestation and/or lactation results in metabolic syndrome in their offspring. Here, we investigated the effect of maternal LP diet during puberty and adulthood on the metabolic homeostasis of glucose and lipids in offspring. Female mice were fed with normal-protein (NP) diet or a LP diet for 11 weeks. Male offspring were then fed with a high-fat diet (NP-HFD and LP-HFD groups) or standard chow diet (NP-Chow and LP-Chow groups) for 4 months. Results showed that maternal LP diet during puberty and adulthood did not alter the insulin sensitivity and hepatic lipid homeostasis of their offspring under chow diet, but aggravated insulin resistance, hepatic steatosis, and hypercholesterolemia of offspring in response to a post-weaning HFD. Accordingly, transcriptomics study with offspring’s liver indicated that several genes related to glucose and lipid metabolism, including lipoprotein lipase (Lpl), long-chain acyl-CoA synthetase 1 (Acsl1), Apoprotein A1 (Apoa1), major urinary protein 19 (Mup19), cholesterol 7α hydroxylase (Cyp7a1) and fibroblast growth factor 1 (Fgf1), were changed by maternal LP diet. Taken together, maternal LP diet during puberty and adulthood could disarrange the expression of metabolic genes in the liver of offspring and aggravate insulin resistance and hepatic steatosis in offspring fed a HFD.
Collapse
Affiliation(s)
- Xiaohua Huang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Yong Zhuo
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Dandan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Yingguo Zhu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhengfeng Fang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lianqiang Che
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Lin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Shengyu Xu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lun Hua
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuanfeng Zou
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Chao Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Lixia Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - De Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
- Correspondence:
| |
Collapse
|
14
|
Liver-specific overexpression of lipoprotein lipase improves glucose metabolism in high-fat diet-fed mice. PLoS One 2022; 17:e0274297. [PMID: 36099304 PMCID: PMC9469954 DOI: 10.1371/journal.pone.0274297] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 08/25/2022] [Indexed: 11/24/2022] Open
Abstract
The liver is the main organ that regulates lipid and glucose metabolism. Ectopic lipid accumulation in the liver impairs insulin sensitivity and glucose metabolism. Lipoprotein lipase (LPL), mainly expressed in the adipose tissue and muscle, is a key enzyme that regulates lipid metabolism via the hydrolysis of triglyceride in chylomicrons and very-low-density lipoproteins. Here, we aimed to investigate whether the suppression level of hepatic lipid accumulation via overexpression of LPL in mouse liver leads to improved metabolism. To overexpress LPL in the liver, we generated an LPL-expressing adenovirus (Ad) vector using an improved Ad vector that exhibited considerably lower hepatotoxicity (Ad-LPL). C57BL/6 mice were treated with Ad vectors and simultaneously fed a high-fat diet (HFD). Lipid droplet formation in the liver decreased in Ad-LPL-treated mice relative to that in control Ad vector-treated mice. Glucose tolerance and insulin resistance were remarkably improved in Ad-LPL-treated mice compared to those in control Ad vector-treated mice. The expression levels of fatty acid oxidation-related genes, such as peroxisome proliferator-activated receptor α, carnitine palmitoyltransferase 1, and acyl-CoA oxidase 1, were 1.7–2.0-fold higher in Ad-LPL-treated mouse livers than that in control Ad-vector-treated mouse livers. Furthermore, hepatic LPL overexpression partly maintained mitochondrial content in HFD-fed mice. These results indicate that LPL overexpression in the livers of HFD-fed mice attenuates the accumulation of lipid droplets in the liver and improves glucose metabolism. These findings may enable the development of new drugs to treat metabolic syndromes such as type 2 diabetes mellitus and non-alcoholic fatty liver disease.
Collapse
|
15
|
Lockridge A, Hanover JA. A nexus of lipid and O-Glcnac metabolism in physiology and disease. Front Endocrinol (Lausanne) 2022; 13:943576. [PMID: 36111295 PMCID: PMC9468787 DOI: 10.3389/fendo.2022.943576] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Although traditionally considered a glucose metabolism-associated modification, the O-linked β-N-Acetylglucosamine (O-GlcNAc) regulatory system interacts extensively with lipids and is required to maintain lipid homeostasis. The enzymes of O-GlcNAc cycling have molecular properties consistent with those expected of broad-spectrum environmental sensors. By direct protein-protein interactions and catalytic modification, O-GlcNAc cycling enzymes may provide both acute and long-term adaptation to stress and other environmental stimuli such as nutrient availability. Depending on the cell type, hyperlipidemia potentiates or depresses O-GlcNAc levels, sometimes biphasically, through a diversity of unique mechanisms that target UDP-GlcNAc synthesis and the availability, activity and substrate selectivity of the glycosylation enzymes, O-GlcNAc Transferase (OGT) and O-GlcNAcase (OGA). At the same time, OGT activity in multiple tissues has been implicated in the homeostatic regulation of systemic lipid uptake, storage and release. Hyperlipidemic patterns of O-GlcNAcylation in these cells are consistent with both transient physiological adaptation and feedback uninhibited obesogenic and metabolic dysregulation. In this review, we summarize the numerous interconnections between lipid and O-GlcNAc metabolism. These links provide insights into how the O-GlcNAc regulatory system may contribute to lipid-associated diseases including obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Amber Lockridge
- Laboratory of Cell and Molecular Biology, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - John A. Hanover
- Laboratory of Cell and Molecular Biology, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
16
|
Deng M, Kutrolli E, Sadewasser A, Michel S, Joibari MM, Jaschinski F, Olivecrona G, Nilsson SK, Kersten S. ANGPTL4 silencing via antisense oligonucleotides reduces plasma triglycerides and glucose in mice without causing lymphadenopathy. J Lipid Res 2022; 63:100237. [PMID: 35667416 PMCID: PMC9270256 DOI: 10.1016/j.jlr.2022.100237] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/30/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022] Open
Abstract
Angiopoietin-like 4 (ANGPTL4) is an important regulator of plasma triglyceride (TG) levels and an attractive pharmacological target for lowering plasma lipids and reducing cardiovascular risk. Here, we aimed to study the efficacy and safety of silencing ANGPTL4 in the livers of mice using hepatocyte-targeting GalNAc-conjugated antisense oligonucleotides (ASOs). Compared with injections with negative control ASO, four injections of two different doses of ANGPTL4 ASO over 2 weeks markedly downregulated ANGPTL4 levels in liver and adipose tissue, which was associated with significantly higher adipose LPL activity and lower plasma TGs in fed and fasted mice, as well as lower plasma glucose levels in fed mice. In separate experiments, injection of two different doses of ANGPTL4 ASO over 20 weeks of high-fat feeding reduced hepatic and adipose ANGPTL4 levels but did not trigger mesenteric lymphadenopathy, an acute phase response, chylous ascites, or any other pathological phenotypes. Compared with mice injected with negative control ASO, mice injected with ANGPTL4 ASO showed reduced food intake, reduced weight gain, and improved glucose tolerance. In addition, they exhibited lower plasma TGs, total cholesterol, LDL-C, glucose, serum amyloid A, and liver TG levels. By contrast, no significant difference in plasma alanine aminotransferase activity was observed. Overall, these data suggest that ASOs targeting ANGPTL4 effectively reduce plasma TG levels in mice without raising major safety concerns.
Collapse
Affiliation(s)
- Mingjuan Deng
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, the Netherlands
| | - Elda Kutrolli
- Lipigon Pharmaceuticals AB, Tvistevägen 48C, 907 36, Umeå, Sweden
| | - Anne Sadewasser
- Secarna Pharmaceuticals GmbH & Co. KG, Am Klopferspitz 19, 82152 Planegg, Germany
| | - Sven Michel
- Secarna Pharmaceuticals GmbH & Co. KG, Am Klopferspitz 19, 82152 Planegg, Germany
| | | | - Frank Jaschinski
- Secarna Pharmaceuticals GmbH & Co. KG, Am Klopferspitz 19, 82152 Planegg, Germany
| | - Gunilla Olivecrona
- Lipigon Pharmaceuticals AB, Tvistevägen 48C, 907 36, Umeå, Sweden; Department of Medical Biosciences, Umeå University, SE-901 87, Umeå, Sweden
| | - Stefan K Nilsson
- Lipigon Pharmaceuticals AB, Tvistevägen 48C, 907 36, Umeå, Sweden
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, the Netherlands.
| |
Collapse
|
17
|
Li H, Liu G, Wan X, Zhou L, Qin ZB, Ma XH, Su K, Liu YJ, Yuan J, Wei CC, Ren AJ, Chen YX, Young SG, Zhang H, Xie Z, Zhang WJ. The zinc finger and BTB domain containing protein ZBTB20 regulates plasma triglyceride metabolism by repressing lipoprotein lipase gene transcription in hepatocytes. Hepatology 2022; 75:1169-1180. [PMID: 34580885 PMCID: PMC9118135 DOI: 10.1002/hep.32176] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Lipoprotein lipase (LPL) is responsible for the lipolytic processing of triglyceride-rich lipoproteins, the deficiency of which causes severe hypertriglyceridemia. Liver LPL expression is high in suckling rodents but relatively low at adulthood. However, the regulatory mechanism and functional significance of liver LPL expression are incompletely understood. We have established the zinc finger protein ZBTB20 as a critical factor for hepatic lipogenesis. Here, we evaluated the role of ZBTB20 in regulating liver Lpl gene transcription and plasma triglyceride metabolism. APPROACH AND RESULTS Hepatocyte-specific inactivation of ZBTB20 in mice led to a remarkable increase in LPL expression at the mRNA and protein levels in adult liver, in which LPL protein was mainly localized onto sinusoidal epithelial cells and Kupffer cells. As a result, the LPL activity in postheparin plasma was substantially increased, and postprandial plasma triglyceride clearance was significantly enhanced, whereas plasma triglyceride levels were decreased. The dysregulated liver LPL expression and low plasma triglyceride levels in ZBTB20-deficient mice were normalized by inactivating hepatic LPL expression. ZBTB20 deficiency protected the mice against high-fat diet-induced hyperlipidemia without causing excessive triglyceride accumulation in the liver. Chromatin immunoprecipitation and gel-shift assay studies revealed that ZBTB20 binds to the LPL promoter in the liver. A luciferase reporter assay revealed that ZBTB20 inhibits the transcriptional activity of LPL promoter. The regulation of LPL expression by ZBTB20 is liver-specific under physiological conditions. CONCLUSIONS Liver ZBTB20 serves as a key regulator of LPL expression and plasma triglyceride metabolism and could be a therapeutic target for hypertriglyceridemia.
Collapse
Affiliation(s)
- Hao Li
- Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Gan Liu
- Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Xiaoqing Wan
- Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Luting Zhou
- Department of Pathophysiology, Naval Medical University, Shanghai, China
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhen-Bang Qin
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin, China
| | - Xian-Hua Ma
- Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Kai Su
- Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Ya-Jin Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin, China
| | - Jinghao Yuan
- Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Chun-Chun Wei
- Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - An-Jing Ren
- Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Yu-Xia Chen
- Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Stephen G. Young
- Departments of Medicine and Human Genetics, University of California, Los Angeles, USA
| | - Hai Zhang
- Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Zhifang Xie
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weiping J. Zhang
- Department of Pathophysiology, Naval Medical University, Shanghai, China
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin, China
| |
Collapse
|
18
|
Kweon SM, Kim G, Jeong Y, Huang W, Lee JS, Lai KKY. Midnolin Regulates Liver Cancer Cell Growth In Vitro and In Vivo. Cancers (Basel) 2022; 14:1421. [PMID: 35326575 PMCID: PMC8946164 DOI: 10.3390/cancers14061421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/25/2022] [Accepted: 03/07/2022] [Indexed: 11/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) ranks worldwide as one of the most lethal cancers. In spite of the vast existing knowledge about HCC, the pathogenesis of HCC is not completely understood. Discovery of novel genes that contribute to HCC pathogenesis will provide new insights for better understanding and treating HCC. The relatively obscure gene midnolin has been studied for over two decades; however, its biological roles are largely unknown. Our study is the first to demonstrate the functional significance of midnolin in HCC/cancer: Midnolin expression correlates with poor prognosis in HCC patients, and suppression of midnolin severely inhibits tumorigenicity of HCC cells in vitro and in mice and disrupts retinoic acid/lipid metabolism in these cells.
Collapse
Affiliation(s)
- Soo-Mi Kweon
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; (S.-M.K.); (G.K.)
| | - Gayeoun Kim
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; (S.-M.K.); (G.K.)
| | - Yunseong Jeong
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.J.); (J.-S.L.)
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA;
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.J.); (J.-S.L.)
| | - Keane K. Y. Lai
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; (S.-M.K.); (G.K.)
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
19
|
Role and mechanism of the action of angiopoietin-like protein ANGPTL4 in plasma lipid metabolism. J Lipid Res 2021; 62:100150. [PMID: 34801488 PMCID: PMC8666355 DOI: 10.1016/j.jlr.2021.100150] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 11/24/2022] Open
Abstract
Triglycerides are carried in the bloodstream as the components of very low-density lipoproteins and chylomicrons. These circulating triglycerides are primarily hydrolyzed in muscle and adipose tissue by the enzyme lipoprotein lipase (LPL). The activity of LPL is regulated by numerous mechanisms, including by three members of the angiopoietin-like protein family: ANGPTL3, ANGPTL4, and ANGPTL8. In this review, we discuss the recent literature concerning the role and mechanism of action of ANGPTL4 in lipid metabolism. ANGPTL4 is a fasting- and lipid-induced factor secreted by numerous cells, including adipocytes, hepatocytes, (cardio)myocytes, and macrophages. In adipocytes, ANGPTL4 mediates the fasting-induced repression of LPL activity by promoting the unfolding of LPL, leading to the cleavage and subsequent degradation of LPL. The inhibition of LPL by ANGPTL4 is opposed by ANGPTL8, which keeps the LPL active after feeding. In macrophages and (cardio)myocytes, ANGPTL4 functions as a lipid-inducible feedback regulator of LPL-mediated lipid uptake. In comparison, in hepatocytes, ANGPTL4 functions as a local inhibitor of hepatic lipase and possibly as an endocrine inhibitor of LPL in extra-hepatic tissues. At the genetic level, loss-of-function mutations in ANGPTL4 are associated with lower plasma triglycerides and higher plasma HDL-C levels, and a reduced risk of coronary artery disease, suggesting that ANGPTL4 is a viable pharmacological target for reducing cardiovascular risk. Whole-body targeting of ANGPTL4 is contraindicated because of severe pathological complications, whereas liver-specific inactivation of ANGPTL4, either as monotherapy or coupled to anti-ANGPTL3 therapies might be a suitable strategy for lowering plasma triglycerides in selected patient groups. In conclusion, the tissue-specific targeting of ANGPTL4 appears to be a viable pharmacological approach to reduce circulating triglycerides.
Collapse
|
20
|
Irungbam K, Roderfeld M, Glimm H, Hempel F, Schneider F, Hehr L, Glebe D, Churin Y, Morlock G, Yüce I, Roeb E. Cholestasis impairs hepatic lipid storage via AMPK and CREB signaling in hepatitis B virus surface protein transgenic mice. J Transl Med 2020; 100:1411-1424. [PMID: 32612285 PMCID: PMC7572243 DOI: 10.1038/s41374-020-0457-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/03/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
Clinical studies demonstrated that nonalcoholic steatohepatitis is associated with liver-related outcomes in chronic hepatitis B. Furthermore, primary biliary fibrosis and biliary atresia occurred in patients with HBV infection. Interestingly, hepatitis B virus surface protein (HBs) transgenic mice spontaneously develop hepatic steatosis. Our aim is to investigate the effect of Abcb4 knockout-induced cholestasis on liver steatosis in HBs transgenic mice. Hybrids of HBs transgenic and Abcb4-/- mice were bred on the BALB/c genetic background. Lipid synthesis, storage, and catabolism as well as proteins and genes that control lipid metabolism were analyzed using HPTLC, qPCR, western blot, electrophoretic mobility shift assay (EMSA), lipid staining, and immunohistochemistry. Hepatic neutral lipid depots were increased in HBs transgenic mice and remarkably reduced in Abcb4-/- and HBs/Abcb4-/- mice. Similarly, HPTLC-based quantification analyses of total hepatic lipid extracts revealed a significant reduction in the amount of triacylglycerols (TAG), while the amount of free fatty acids (FFA) was increased in Abcb4-/- and HBs/Abcb4-/- in comparison to wild-type and HBs mice. PLIN2, a lipid droplet-associated protein, was less expressed in Abcb4-/- and HBs/Abcb4-/-. The expression of genes-encoding proteins involved in TAG synthesis and de novo lipogenesis (Agpat1, Gpat1, Mgat1, Dgat1, Dgat2, Fasn, Hmgcs1, Acc1, Srebp1-c, and Pparγ) was suppressed, and AMPK and CREB were activated in Abcb4-/- and HBs/Abcb4-/- compared to wild-type and HBs mice. Simulating cholestatic conditions in cell culture resulted in AMPK and CREB activation while FASN and PLIN2 were reduced. A concurrent inhibition of AMPK signaling revealed normal expression level of FASN and PLIN2, suggesting that activation of AMPK-CREB signaling regulates hepatic lipid metabolism, i.e. synthesis and storage, under cholestatic condition. In conclusions, in vivo and mechanistic in vitro data suggest that cholestasis reduces hepatic lipid storage via AMPK and CREB signaling. The results of the current study could be the basis for novel therapeutic strategies as NASH is a crucial factor that can aggravate chronic liver diseases.
Collapse
Affiliation(s)
- Karuna Irungbam
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Martin Roderfeld
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Hannah Glimm
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Felix Hempel
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Franziska Schneider
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Laura Hehr
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Dieter Glebe
- Institute of Medical Virology, National Reference Centre for Hepatitis B Viruses and Hepatitis D Viruses, Justus Liebig University, Giessen, Germany
| | - Yuri Churin
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Gertrud Morlock
- Institute of Nutritional Science, Chair of Food Science, and TransMIT Center for Effect-Directed Analysis, Justus Liebig University Giessen, Giessen, Germany
| | - Imanuel Yüce
- Institute of Nutritional Science, Chair of Food Science, and TransMIT Center for Effect-Directed Analysis, Justus Liebig University Giessen, Giessen, Germany
| | - Elke Roeb
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany.
| |
Collapse
|
21
|
Barchetta I, Cimini FA, Chiappetta C, Bertoccini L, Ceccarelli V, Capoccia D, Gaggini M, Di Cristofano C, Della Rocca C, Silecchia G, Leonetti F, Lenzi A, Gastaldelli A, Cavallo MG. Relationship between hepatic and systemic angiopoietin-like 3, hepatic Vitamin D receptor expression and NAFLD in obesity. Liver Int 2020; 40:2139-2147. [PMID: 32510837 DOI: 10.1111/liv.14554] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide and an independent risk factor for cardiovascular mortality. Angiopoietin-like proteins (ANGPTLs) are targets for vitamin D receptor (VDR)-mediated gene transcription and this axis may promote NAFLD. ANGPTL3 is a hepatokine which inhibits lipoprotein lipase and its experimentally induced inactivation reduces hepatosteatosis. Little is known on ANGPTL3 in human NAFLD and no data exist on its relationship with hepatic VDR/VD-related genes. The aim of this research was to investigate hepatic ANGPTLs and VDR/VD-related gene expression in human obesity in relation to NAFLD. METHODS We conducted a cross-sectional investigation on forty obese subjects with/without NAFLD. We evaluated hepatic ANGPTL3, ANGPTL4, ANGPTL8, LPL, VDR, CYP27A1 and CYP2R1 mRNA expression in liver biopsies by RT-PCR; VDR expression was further investigated by immunohistochemistry; circulating ANGPTL3 was measured by Milliplex assay. RESULTS Compared to non-NAFLD, NAFLD individuals had significantly higher hepatic VDR, ANGPTL3 and LPL expression. ANGPTL3 correlated with steatosis grade, LPL, VDR, CYP27A1 and CYP2R1 expression. Plasma ANGPTL3 concentrations were positively associated with clinical/histological markers of NAFLD/NASH and with hepatic ANGPTL3 expression. Greater hepatic VDR expression was the main determinant of hepatic ANGPTL3 after adjusting for multiple confounders. CONCLUSIONS Hepatic ANGPTL3 expression correlates with greater VDR expression, presence and severity of NAFLD and translates in increased circulating ANGPTL3, likely as a result of its modulation by up-regulated hepatic VDR in NAFLD. This study provides novel insights to potential mechanisms underlying ANGPTLs-mediated ectopic fat accumulation and NAFLD development in obesity.
Collapse
Affiliation(s)
- Ilaria Barchetta
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Flavia A Cimini
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Caterina Chiappetta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Laura Bertoccini
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Danila Capoccia
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Melania Gaggini
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, Pisa, Italy
| | - Claudio Di Cristofano
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Carlo Della Rocca
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Gianfranco Silecchia
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Frida Leonetti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, Pisa, Italy
| | - Maria G Cavallo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
22
|
El Hage R, Hernandez-Sanabria E, Calatayud Arroyo M, Van de Wiele T. Supplementation of a propionate-producing consortium improves markers of insulin resistance in an in vitro model of gut-liver axis. Am J Physiol Endocrinol Metab 2020; 318:E742-E749. [PMID: 31935110 DOI: 10.1152/ajpendo.00523.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Gut-liver cross talk is an important determinant of human health with profound effects on energy homeostasis. While gut microbes produce a huge range of metabolites, specific compounds such as short-chain fatty acids (SCFAs) can enter the portal circulation and reach the liver (Brandl K, Schnabl B. Curr Opin Gastroenterol 33: 128-133, 2017), a central organ involved in glucose homeostasis and diabetes control. Propionate is a major SCFA involved in activation of intestinal gluconeogenesis (IGN), thereby regulating food intake, enhancing insulin sensitivity, and leading to metabolic homeostasis. Although microbiome-modulating strategies may target the increased microbial production of propionate, it is not clear whether such an effect spreads through to the hepatic cellular level. Here, we designed a propionate-producing consortium using a selection of commensal gut bacteria, and we investigated how their delivered metabolites impact an in vitro enterohepatic model of insulin resistance. Glycogen storage on hepatocyte-like cells and inflammatory markers associated with insulin resistance were evaluated to understand the role of gut metabolites on gut-liver cross talk in a simulated scenario of insulin resistance. The metabolites produced by our consortium increased glycogen synthesis by ~57% and decreased proinflammatory markers such as IL-8 by 12%, thus elucidating the positive effect of our consortium on metabolic function and low-grade inflammation. Our results suggest that microbiota-derived products can be a promising multipurpose strategy to modulate energy homeostasis, with the potential ability to assist in managing metabolic diseases due to their adaptability.
Collapse
Affiliation(s)
- Racha El Hage
- Center for Microbial Ecology and Technology (CMET), Ghent University, Ghent, Belgium
| | | | | | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Ghent University, Ghent, Belgium
| |
Collapse
|
23
|
Cirera S, Taşöz E, Juul Jacobsen M, Schumacher-Petersen C, Østergaard Christoffersen B, Kaae Kirk R, Pagh Ludvigsen T, Hvid H, Duelund Pedersen H, Høier Olsen L, Fredholm M. The expression signatures in liver and adipose tissue from obese Göttingen Minipigs reveal a predisposition for healthy fat accumulation. Nutr Diabetes 2020; 10:9. [PMID: 32205840 PMCID: PMC7090036 DOI: 10.1038/s41387-020-0112-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Model animals are valuable resources for dissecting basic aspects of the regulation of obesity and metabolism. The translatability of results relies on understanding comparative aspects of molecular pathophysiology. Several studies have shown that despite the presence of overt obesity and dyslipidemia in the pig key human pathological hepatic findings such as hepatocellular ballooning and abundant steatosis are lacking in the model. OBJECTIVES The aim of this study was to elucidate why these histopathological characteristics did not occur in a high fat, fructose and cholesterol (FFC) diet-induced obese Göttingen Minipig model. METHODS High-throughput expression profiling of more than 90 metabolically relevant genes was performed in liver, subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) of male minipigs diet fed: standard chow (SD, n = 7); FFC diet (n = 14); FFC diet in streptozotocin-induced diabetic pigs (FFCDIA, n = 8). Moreover, histopathological assessment of SAT and VAT was performed. RESULTS 12, 4 and 1 genes were highly significantly differentially expressed in liver, SAT and VAT when comparing the FFC and SD groups whereas the corresponding numbers were 15, 2, and 1 when comparing the FFCDIA and SD groups. Although the minipigs in both FFC groups developed sever obesity and dyslipidemia, the insulin-signaling pathways were not affected. Notably, four genes involved in lipid acquisition and removal, were highly deregulated in the liver: PPARG, LPL, CD36 and FABP4. These genes have been reported to play a major role in promoting hepatic steatosis in rodents and humans. Since very little macrophage-associated pro-inflammatory response was detected in the adipose tissues the expansion appears to have no adverse impact on adipose tissue metabolism. CONCLUSION The study shows that morbidly obese Göttingen Minipigs are protected against many of the metabolic and hepatic abnormalities associated with obesity due to a remarkable ability to expand the adipose compartments to accommodate excess calories.
Collapse
Affiliation(s)
- Susanna Cirera
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | - Emirhan Taşöz
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | - Mette Juul Jacobsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | - Camilla Schumacher-Petersen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | | | - Rikke Kaae Kirk
- Global Drug Discovery, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | | | - Henning Hvid
- Global Drug Discovery, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - Henrik Duelund Pedersen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
- Ellegaard Gottingen Minipigs A/S, Sorø Landevej 302, 4261, Dalmose, Denmark
| | - Lisbeth Høier Olsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | - Merete Fredholm
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark.
| |
Collapse
|
24
|
Nadermann N, Volkoff H. Effects of short-term exercise on food intake and the expression of appetite-regulating factors in goldfish. Peptides 2020; 123:170182. [PMID: 31678371 DOI: 10.1016/j.peptides.2019.170182] [Citation(s) in RCA: 4] [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: 08/15/2019] [Revised: 10/15/2019] [Accepted: 10/23/2019] [Indexed: 12/30/2022]
Abstract
In mammals, growing evidence indicates that exercise affects food intake, metabolism and the expression and blood levels of appetite regulators. In this study, we examined the effects of short-term (30 min, at low and high water flow) exercise on food intake, glucose levels and the expressions of appetite regulators in goldfish hypothalamus (irisin, orexin, CART, leptin), intestine (CCK, PYY, proglucagon/GLP-1), muscle (irisin) and liver (leptin), of brain-derived neurotrophic factor (BDNF) in brain, interleukin-6 (IL6) in muscle and hypothalamus, and major metabolic enzymes, the glycolytic enzyme glucokinase (GCK) and its regulatory protein (GCKR) in liver, the lipolytic enzyme lipoprotein lipase in intestine and muscle, and trypsin in intestine. Fish submitted to high flow exercise had a lower post-exercise food intake compared to control fish but no differences were seen in glucose levels between groups. Exercise induced an increase in hypothalamic expression levels of CART, IL6 and BDNF, but not orexin, irisin, CRF, leptin and NPY. High flow exercise induced an increase in intestine CCK, PYY and GLP-1, and muscle irisin and IL-6 expression levels. Exercise had no effects on expression levels of hepatic leptin or any of the metabolic enzymes examined. Our results suggest that, in goldfish, short-term exercise might decrease feeding in part by affecting the expressions of myokines and peripheral, but not central appetite regulators or metabolic enzyme/hormones.
Collapse
Affiliation(s)
- Noelle Nadermann
- Departments of Biology and Biochemistry, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada; Hochschule Mannheim University, Mannheim, 68163, Germany
| | - Hélène Volkoff
- Departments of Biology and Biochemistry, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada.
| |
Collapse
|
25
|
Bhushan B, Banerjee S, Paranjpe S, Koral K, Mars WM, Stoops JW, Orr A, Bowen WC, Locker J, Michalopoulos GK. Pharmacologic Inhibition of Epidermal Growth Factor Receptor Suppresses Nonalcoholic Fatty Liver Disease in a Murine Fast-Food Diet Model. Hepatology 2019; 70:1546-1563. [PMID: 31063640 DOI: 10.1002/hep.30696] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 04/29/2019] [Indexed: 12/13/2022]
Abstract
Epidermal growth factor receptor (EGFR) is a critical regulator of hepatocyte proliferation and liver regeneration. Our recent work indicated that EGFR can also regulate lipid metabolism during liver regeneration after partial hepatectomy. Based on these findings, we investigated the role of EGFR in a mouse model of nonalcoholic fatty liver disease (NAFLD) using a pharmacological inhibition strategy. C57BL6/J mice were fed a chow diet or a fast-food diet (FFD) with or without EGFR inhibitor (canertinib) for 2 months. EGFR inhibition completely prevented development of steatosis and liver injury in this model. In order to study if EGFR inhibition can reverse NAFLD progression, mice were fed the FFD for 5 months, with or without canertinib treatment for the last 5 weeks of the study. EGFR inhibition remarkably decreased steatosis, liver injury, and fibrosis and improved glucose tolerance. Microarray analysis revealed that ~40% of genes altered by the FFD were differentially expressed after EGFR inhibition and, thus, are potentially regulated by EGFR. Several genes and enzymes related to lipid metabolism (particularly fatty acid synthesis and lipolysis), which were disrupted by the FFD, were found to be modulated by EGFR. Several crucial transcription factors that play a central role in regulating these lipid metabolism genes during NAFLD, including peroxisome proliferator-activated receptor gamma (PPARγ), sterol regulatory element-binding transcription factor 1 (SREBF1), carbohydrate-responsive element-binding protein, and hepatocyte nuclear factor 4 alpha, were also found to be modulated by EGFR. In fact, chromatin immunoprecipitation analysis revealed that PPARγ binding to several crucial lipid metabolism genes (fatty acid synthase, stearoyl-coenzyme A desaturase 1, and perilipin 2) was drastically reduced by EGFR inhibition. Further upstream, EGFR inhibition suppressed AKT signaling, which is known to control these transcription factors, including PPARγ and SREBF1, in NAFLD models. Lastly, the effect of EGFR in FFD-induced fatty-liver phenotype was not shared by receptor tyrosine kinase MET, investigated using MET knockout mice. Conclusion: Our study revealed a role of EGFR in NAFLD and the potential of EGFR inhibition as a treatment strategy for NAFLD.
Collapse
Affiliation(s)
- Bharat Bhushan
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Swati Banerjee
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Shirish Paranjpe
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Kelly Koral
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Wendy M Mars
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - John W Stoops
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Anne Orr
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - William C Bowen
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Joseph Locker
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | | |
Collapse
|
26
|
Kang I, Park M, Yang SJ, Lee M. Lipoprotein Lipase Inhibitor, Nordihydroguaiaretic Acid, Aggravates Metabolic Phenotypes and Alters HDL Particle Size in the Western Diet-Fed db/db Mice. Int J Mol Sci 2019; 20:ijms20123057. [PMID: 31234537 PMCID: PMC6627211 DOI: 10.3390/ijms20123057] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 12/19/2022] Open
Abstract
Lipoprotein lipase (LPL) hydrolyzes triglycerides in lipoprotein to supply fatty acids, and its deficiency leads to hypertriglyceridemia, thereby inducing metabolic syndrome (MetSyn). Nordihydroguaiaretic acid (NDGA) has been recently reported to inhibit LPL secretion by endoplasmic reticulum (ER)-Golgi redistribution. However, the role of NDGA on dyslipidemia and MetSyn remains unclear. To address this question, leptin receptor knock out (KO)-db/db mice were randomly assigned to three different groups: A normal AIN76-A diet (CON), a Western diet (WD) and a Western diet with 0.1% NDGA and an LPL inhibitor, (WD+NDGA). All mice were fed for 12 weeks. The LPL inhibition by NDGA was confirmed by measuring the systemic LPL mass and adipose LPL gene expression. We investigated whether the LPL inhibition by NDGA alters the metabolic phenotypes. NDGA led to hyperglycemia, hypertriglyceridemia, and hypercholesterolemia. More strikingly, the supplementation of NDGA increased the percentage of high density lipoprotein (HDL)small (HDL3a+3b+3c) and decreased the percentage of HDLlarge (HDL2a+2b) compared to the WD group, which indicates that LPL inhibition modulates HDL subclasses. was NDGA increased adipose inflammation but had no impact on hepatic stress signals. Taken together, these findings demonstrated that LPL inhibition by NDGA aggravates metabolic parameters and alters HDL particle size.
Collapse
Affiliation(s)
- Inhae Kang
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Korea.
| | - Miyoung Park
- Research Institute of Obesity Sciences, Sungshin Women's University, Seoul 01133, Korea.
| | - Soo Jin Yang
- Department of Food and Nutrition, Seoul Women's University, Seoul 01797, Korea.
| | - Myoungsook Lee
- Research Institute of Obesity Sciences, Sungshin Women's University, Seoul 01133, Korea.
- Department of Food and Nutrition, Sungshin Women's University, Seoul 01133, Korea.
| |
Collapse
|
27
|
Kshatriya D, Li X, Giunta GM, Yuan B, Zhao D, Simon JE, Wu Q, Bello NT. Phenolic-enriched raspberry fruit extract (Rubus idaeus) resulted in lower weight gain, increased ambulatory activity, and elevated hepatic lipoprotein lipase and heme oxygenase-1 expression in male mice fed a high-fat diet. Nutr Res 2019; 68:19-33. [PMID: 31252376 DOI: 10.1016/j.nutres.2019.05.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/06/2019] [Accepted: 05/17/2019] [Indexed: 12/15/2022]
Abstract
Red raspberries (Rubus idaeus) contain numerous phenolic compounds with purported health benefits. Raspberry ketone (4-(4-hydroxyphenyl)-2-butanone) is a primary raspberry flavor phenolic found in raspberries and is designated as a synthetic flavoring agent by the Food and Drug Administration. Synthetic raspberry ketone has been demonstrated to result in weight loss in rodents. We tested whether phenolic-enriched raspberry extracts, compared with raspberry ketone, would be more resilient to the metabolic alterations caused by an obesogenic diet. Male C57BL/6J mice (8 weeks old) received a daily oral dose of vehicle (VEH; 50% propylene glycol, 40% water, and 10% dimethyl sulfoxide), raspberry extract low (REL; 0.2 g/kg), raspberry extract high (REH; 2 g/kg), or raspberry ketone (RK; 0.2 g/kg). Coincident with daily dosing, mice were placed on a high-fat diet (45% fat). After 4 weeks, REH and RK reduced body weight gain (approximately 5%-9%) and white adipose mass (approximately 20%) compared with VEH. Hepatic gene expression of heme oxygenase-1 and lipoprotein lipase was upregulated in REH compared with VEH. Indirect calorimetry indicated that respiratory exchange ratio (CO2 production to O2 consumption) was lower, suggesting increased fat oxidation with all treatments. REH treatment increased total ambulatory behavior. Energy expenditure/lean mass was higher in REH compared with REL treatment. There were no treatment differences in cumulative intake, meal patterns, or hypothalamic feed-related gene expression. Our results suggest that raspberry ketone and a phenolic-enriched raspberry extract both have the capacity to prevent weight gain but differ in the preventative mechanisms for excess fat accumulation following high-fat diet exposure.
Collapse
Affiliation(s)
- Dushyant Kshatriya
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA; Nutritional Sciences Graduate Program, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey New Brunswick, NJ, 08901, USA
| | - Xinyi Li
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA; Nutritional Sciences Graduate Program, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey New Brunswick, NJ, 08901, USA
| | - Gina M Giunta
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Bo Yuan
- New Use Agriculture and Natural Plant Products Program, Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Danyue Zhao
- New Use Agriculture and Natural Plant Products Program, Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - James E Simon
- New Use Agriculture and Natural Plant Products Program, Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Qingli Wu
- New Use Agriculture and Natural Plant Products Program, Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Nicholas T Bello
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA; Nutritional Sciences Graduate Program, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey New Brunswick, NJ, 08901, USA.
| |
Collapse
|
28
|
Allan CM, Heizer PJ, Tu Y, Sandoval NP, Jung RS, Morales JE, Sajti E, Troutman TD, Saunders TL, Cusanovich DA, Beigneux AP, Romanoski CE, Fong LG, Young SG. An upstream enhancer regulates Gpihbp1 expression in a tissue-specific manner. J Lipid Res 2019; 60:869-879. [PMID: 30598475 PMCID: PMC6446700 DOI: 10.1194/jlr.m091322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/02/2018] [Indexed: 01/22/2023] Open
Abstract
Glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1), the protein that shuttles LPL to the capillary lumen, is essential for plasma triglyceride metabolism. When GPIHBP1 is absent, LPL remains stranded within the interstitial spaces and plasma triglyceride hydrolysis is impaired, resulting in severe hypertriglyceridemia. While the functions of GPIHBP1 in intravascular lipolysis are reasonably well understood, no one has yet identified DNA sequences regulating GPIHBP1 expression. In the current studies, we identified an enhancer element located ∼3.6 kb upstream from exon 1 of mouse Gpihbp1. To examine the importance of the enhancer, we used CRISPR/Cas9 genome editing to create mice lacking the enhancer (Gpihbp1Enh/Enh). Removing the enhancer reduced Gpihbp1 expression by >90% in the liver and by ∼50% in heart and brown adipose tissue. The reduced expression of GPIHBP1 was insufficient to prevent LPL from reaching the capillary lumen, and it did not lead to hypertriglyceridemia-even when mice were fed a high-fat diet. Compound heterozygotes (Gpihbp1Enh/- mice) displayed further reductions in Gpihbp1 expression and exhibited partial mislocalization of LPL (increased amounts of LPL within the interstitial spaces of the heart), but the plasma triglyceride levels were not perturbed. The enhancer element that we identified represents the first insight into DNA sequences controlling Gpihbp1 expression.
Collapse
Affiliation(s)
- Christopher M Allan
- Departments of Medicine University of California, Los Angeles, Los Angeles, CA 90095
| | - Patrick J Heizer
- Departments of Medicine University of California, Los Angeles, Los Angeles, CA 90095
| | - Yiping Tu
- Departments of Medicine University of California, Los Angeles, Los Angeles, CA 90095
| | - Norma P Sandoval
- Departments of Medicine University of California, Los Angeles, Los Angeles, CA 90095
| | - Rachel S Jung
- Departments of Medicine University of California, Los Angeles, Los Angeles, CA 90095
| | - Jazmin E Morales
- Departments of Medicine University of California, Los Angeles, Los Angeles, CA 90095
| | - Eniko Sajti
- Department of Pediatrics, Division of Neurology, Rady Children's Hospital, University of California, San Diego, San Diego, CA 92123
| | - Ty D Troutman
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Thomas L Saunders
- University of Michigan Transgenic Animal Model Core, Department of Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Darren A Cusanovich
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721
| | - Anne P Beigneux
- Departments of Medicine University of California, Los Angeles, Los Angeles, CA 90095
| | - Casey E Romanoski
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721.
| | - Loren G Fong
- Departments of Medicine University of California, Los Angeles, Los Angeles, CA 90095.
| | - Stephen G Young
- Departments of Medicine University of California, Los Angeles, Los Angeles, CA 90095; Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095.
| |
Collapse
|
29
|
Octacosanol and policosanol prevent high-fat diet-induced obesity and metabolic disorders by activating brown adipose tissue and improving liver metabolism. Sci Rep 2019; 9:5169. [PMID: 30914769 PMCID: PMC6435753 DOI: 10.1038/s41598-019-41631-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/13/2019] [Indexed: 01/02/2023] Open
Abstract
Brown adipose tissue (BAT) is an attractive therapeutic target for treating obesity and metabolic diseases. Octacosanol is the main component of policosanol, a mixture of very long chain aliphatic alcohols obtained from plants. The current study aimed to investigate the effect of octacosanol and policosanol on high-fat diet (HFD)-induced obesity. Mice were fed on chow, or HFD, with or without octacosanol or policosanol treatment for four weeks. HFD-fed mice showed significantly higher body weight and body fat compared with chow-fed mice. However, mice fed on HFD treated with octacosanol or policosanol (HFDo/p) showed lower body weight gain, body fat gain, insulin resistance and hepatic lipid content. Lower body fat gain after octacosanol or policosanol was associated with increased BAT activity, reduced expression of genes involved in lipogenesis and cholesterol uptake in the liver, and amelioration of white adipose tissue (WAT) inflammation. Moreover, octacosanol and policosanol significantly increased the expression of Ffar4, a gene encoding polyunsaturated fatty acid receptor, which activates BAT thermogenesis. Together, these results suggest that octacosanol and policosanol ameliorate diet-induced obesity and metabolic disorders by increasing BAT activity and improving hepatic lipid metabolism. Thus, these lipids represent promising therapeutic targets for the prevention and treatment of obesity and obesity-related metabolic disorders.
Collapse
|
30
|
Sendi H, Mead I, Wan M, Mehrab-Mohseni M, Koch K, Atala A, Bonkovsky HL, Bishop CE. miR-122 inhibition in a human liver organoid model leads to liver inflammation, necrosis, steatofibrosis and dysregulated insulin signaling. PLoS One 2018; 13:e0200847. [PMID: 30024933 PMCID: PMC6053181 DOI: 10.1371/journal.pone.0200847] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/03/2018] [Indexed: 12/12/2022] Open
Abstract
To investigate the role of miR-122 in the development and regression of non-alcoholic fatty liver disease (NAFLD) in vitro, we used multicellular 3D human liver organoids developed in our laboratory. These organoids consist of primary human hepatocytes, Kupffer cells, quiescent stellate cells and liver sinusoidal endothelial cells. They remain viable and functional for 4 weeks expressing typical markers of liver function such as synthesis of albumin, urea, and alpha-1 p450 drug metabolism. Before mixing, hepatic cells were transduced with lentivirus to inhibit miR122 expression (ABM, CA). Immediately after the organoids were fully formed (day 4) or after 1 or 2 weeks of additional incubation (days 11 or 18), the organoids were analyzed using fluorescent live/dead staining and ATP production; total RNA was extracted for qPCR gene expression profiling. Our results show that miR-122 inhibition in liver organoids leads to inflammation, necrosis, steatosis and fibrosis. This was associated with increase in inflammatory cytokines (IL6, TNF), chemokines (CCL2, CCL3) and increase in a subset of Matrix Metaloproteinases (MMP8, MMP9). An altered expression of key genes in lipid metabolism (i.e LPL, LDLR) and insulin signaling (i.e GLUT4, IRS1) was also identified. Conclusion: Our results highlight the role of miR-122 inhibition in liver inflammation, steatofibrosis and dysregulation of insulin signaling. Patients with NAFLD are known to have altered levels of miR-122, therefore we suggest that miR-122 mimics could play a useful role in reversing liver steatofibrosis and insulin resistance seen in patients with NAFLD.
Collapse
Affiliation(s)
- Hossein Sendi
- The Laboratory for Liver Diseases and Metabolic Disorders, Section on Gastroenterology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
- * E-mail:
| | - Ivy Mead
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
| | - Meimei Wan
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
| | - Marjan Mehrab-Mohseni
- The Laboratory for Liver Diseases and Metabolic Disorders, Section on Gastroenterology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Kenneth Koch
- The Laboratory for Liver Diseases and Metabolic Disorders, Section on Gastroenterology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Anthony Atala
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
| | - Herbert L. Bonkovsky
- The Laboratory for Liver Diseases and Metabolic Disorders, Section on Gastroenterology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Colin E. Bishop
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
| |
Collapse
|
31
|
Hatefi Z, Soltani G, Khosravi S, Kazemi M, Salehi AR, Salehi R. Micro R-410 Binding Site Single Nucleotide Polymorphism rs13702 in Lipoprotein Lipase Gene is Effective to Increase Susceptibility to Type 2 Diabetes in Iranian Population. Adv Biomed Res 2018; 7:79. [PMID: 29930919 PMCID: PMC5991288 DOI: 10.4103/abr.abr_286_16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The relationship between dyslipidemia and type 2 diabetes mellitus (T2DM) has been frequently reported. Lipoprotein lipase (LPL) is considered to be an effective gene in regulating lipid profile. MicroRNAs (miRNAs) are small noncoding RNAs involved in posttranscriptional regulation of gene expression. In the present study, we have evaluated rs13702 (C/T) polymorphism located in miRNA-410 binding site of LPL gene in subset of Iranian T2DM patients and their normal counterparts. MATERIALS AND METHODS In this case-control study, 102 T2DM patients and 98 healthy controls were worked out for rs13702 single nucleotide polymorphism genotypes. High resolution meting (HRM) analysis was used for genotyping. RESULTS C allele of rs13702 C/T polymorphism located in miRNA-410 binding site in LPL gene was detected to be significantly associated with T2DM (C allele; odds ratios (OR) = 1.729 (95% confidential intervals (CI) = 1.184-2.523); P = 0.005) also its CC genotype (OR = 3.28 (95% CI 8.68-1.24); P = 0.010) showed the same association. CONCLUSION Correlation of rs13702 C allele with susceptibility to T2DM may be due to the higher level of LPL that leads to increased plasma fatty acids and its entry into peripheral tissues such as skeletal muscle, liver, and adipocytes causing development of insulin resistance and ultimately T2DM.
Collapse
Affiliation(s)
- Zahra Hatefi
- From the Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Goljahan Soltani
- From the Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sharifeh Khosravi
- From the Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Kazemi
- From the Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ahmad Reza Salehi
- From the Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rasoul Salehi
- From the Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Gerfa Namayesh Azmayesh (GENAZMA) Science and Research Institute, Isfahan, Iran
| |
Collapse
|
32
|
Chen Q, Niu L, Hua C, Geng Y, Cai L, Tao S, Ni Y, Zhao R. Chronic dexamethasone exposure markedly decreased the hepatic triglyceride accumulation in growing goats. Gen Comp Endocrinol 2018; 259:115-121. [PMID: 29155266 DOI: 10.1016/j.ygcen.2017.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/05/2017] [Accepted: 11/15/2017] [Indexed: 02/07/2023]
Abstract
Chronic stress seriously threatens welfare and health in animals and humans. Consecutive dexamethasone (Dex) injection was used to mimic chronic stress previously. In order to investigate the effect of chronic stress on hepatic lipids metabolism, in this study, 10 healthy male goats were randomly allocated into two groups, one received a consecutive injection of Dex via intramuscularly for 3 weeks (Dex group), the other received the same volume of saline as the control group (Con group). Hepatic health and triglyceride (TG) metabolism were analyzed and compared between two groups. The data showed that a significant decrease of TG in plasma and the liver was significantly decreased by Dex (P < .05), while the hepatic nonesterified fatty acid (NEFA) concentration was increased compared to the Con group (P < .05). Consistent with the decrease of TG level, the activity of hepatic lipoprotein lipase (LPL) and hepatic lipase (HL) enzymes activities were significantly enhanced by Dex. Real-time PCR results showed that the mRNA expression of sterol regulatory element binding transcription factor 1 (SREBP-1), acyl-CoA dehydrogenase long chain (ACADL) and acyl-CoA synthetase bubblegum family member 1 (ACSBG1) genes in liver was significantly up-regulated by chronic Dex injection (P < .05), whereas perilipin 2 (PLIN2) and adipose triglyceride lipase (ATGL) mRNA expression was significantly decreased by Dex (P < .05). In addition, no obvious damages were observed in the liver in both Con and Dex groups demonstrating by the sirius red staining, HE staining as well as several biochemical parameters related to the functional status of hepatocytes. Our data indicate that chronic Dex exposure decreases TG levels in the circulation and the liver through activating lipolysis and inhibiting lipogenesis without causing hepatic damages in the growing goats.
Collapse
Affiliation(s)
- Qu Chen
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Liqiong Niu
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Canfeng Hua
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yali Geng
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Liuping Cai
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Shiyu Tao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yingdong Ni
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| |
Collapse
|
33
|
Kachaylo E, Tschuor C, Calo N, Borgeaud N, Ungethüm U, Limani P, Piguet AC, Dufour JF, Foti M, Graf R, Clavien PA, Humar B. PTEN Down-Regulation Promotes β-Oxidation to Fuel Hypertrophic Liver Growth After Hepatectomy in Mice. Hepatology 2017; 66:908-921. [PMID: 28437835 DOI: 10.1002/hep.29226] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 03/17/2017] [Accepted: 04/14/2017] [Indexed: 12/25/2022]
Abstract
UNLABELLED In regenerating liver, hepatocytes accumulate lipids before the major wave of parenchymal growth. This transient, regeneration-associated steatosis (TRAS) is required for liver recovery, but its purpose is unclear. The tumor suppressor phosphatase and tensin homolog (PTEN) is a key inhibitor of the protein kinase B/mammalian target of rapamycin axis that regulates growth and metabolic adaptations after hepatectomy. In quiescent liver, PTEN causes pathological steatosis when lost, whereas its role in regenerating liver remains unknown. Here, we show that PTEN down-regulation promotes liver growth in a TRAS-dependent way. In wild-type mice, PTEN reduction occurred after TRAS formation, persisted during its disappearance, and correlated with up-regulated β-oxidation at the expense of lipogenesis. Pharmacological modulation revealed an association of PTEN with TRAS turnover and hypertrophic liver growth. In liver-specific Pten-/- mice shortly after induction of knockout, hypertrophic regeneration was accelerated and led to hepatomegaly. The resulting surplus liver mass was functional, as demonstrated by raised survival in a lethal model of resection-induced liver failure. Indirect calorimetry revealed lipid oxidation as the primary energy source early after hepatectomy. The shift from glucose to lipid usage was pronounced in Pten-/- mice and correlated with the disappearance of TRAS. Partial inhibition of β-oxidation led to persisting TRAS in Pten-/- mice and abrogated hypertrophic liver growth. PTEN down-regulation may promote β-oxidation through β-catenin, whereas hypertrophy was dependent on mammalian target of rapamycin complex 1. CONCLUSION PTEN down-regulation after hepatectomy promotes the burning of TRAS-derived lipids to fuel hypertrophic liver regeneration. Therefore, the anabolic function of PTEN deficiency in resting liver is transformed into catabolic activities upon tissue loss. These findings portray PTEN as a node coordinating liver growth with its energy demands and emphasize the need of lipids for regeneration. (Hepatology 2017;66:908-921).
Collapse
Affiliation(s)
- Ekaterina Kachaylo
- Department of Surgery, Swiss Hepato-Pancreato-Biliary and Transplantation Center, University Hospital Zürich, Zürich, Switzerland
| | - Christoph Tschuor
- Department of Surgery, Swiss Hepato-Pancreato-Biliary and Transplantation Center, University Hospital Zürich, Zürich, Switzerland
| | - Nicolas Calo
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Nathalie Borgeaud
- Department of Surgery, Swiss Hepato-Pancreato-Biliary and Transplantation Center, University Hospital Zürich, Zürich, Switzerland
| | - Udo Ungethüm
- Department of Surgery, Swiss Hepato-Pancreato-Biliary and Transplantation Center, University Hospital Zürich, Zürich, Switzerland
| | - Perparim Limani
- Department of Surgery, Swiss Hepato-Pancreato-Biliary and Transplantation Center, University Hospital Zürich, Zürich, Switzerland
| | - Anne-Christine Piguet
- Hepatology, Department of Clinical Research, University of Berne, Berne, Switzerland
| | - Jean-Francois Dufour
- Hepatology, Department of Clinical Research, University of Berne, Berne, Switzerland
| | - Michelangelo Foti
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Rolf Graf
- Department of Surgery, Swiss Hepato-Pancreato-Biliary and Transplantation Center, University Hospital Zürich, Zürich, Switzerland
| | - Pierre A Clavien
- Department of Surgery, Swiss Hepato-Pancreato-Biliary and Transplantation Center, University Hospital Zürich, Zürich, Switzerland
| | - Bostjan Humar
- Department of Surgery, Swiss Hepato-Pancreato-Biliary and Transplantation Center, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
34
|
Kanaki M, Tiniakou I, Thymiakou E, Kardassis D. Physical and functional interactions between nuclear receptor LXRα and the forkhead box transcription factor FOXA2 regulate the response of the human lipoprotein lipase gene to oxysterols in hepatic cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:848-860. [PMID: 28576574 DOI: 10.1016/j.bbagrm.2017.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/17/2017] [Accepted: 05/29/2017] [Indexed: 11/30/2022]
Abstract
Lipoprotein lipase (LPL) catalyzes the hydrolysis of triglycerides from triglyceride-rich lipoproteins such as VLDL and chylomicrons in the circulation. Mutations in LPL or its activator apolipoprotein C-II cause hypertriglyceridemia in humans and animal models. The levels of LPL in the liver are low but they can be strongly induced by a high cholesterol diet or by synthetic ligands of Liver X Receptors (LXRs). However, the mechanism by which LXRs activate the human LPL gene is unknown. In the present study we show that LXR agonists increased the mRNA and protein levels as well as the promoter activity of human LPL in HepG2 cells. A promoter deletion analysis defined the proximal -109/-28 region, which contains a functional FOXA2 element, as essential for transactivation by ligand-activated LXRα/RXRα heterodimers. Silencing of endogenous FOXA2 in HepG2 cells by siRNAs or by treatment with insulin compromised the induction of the LPL gene by LXR agonists whereas mutations in the FOXA2 site abolished the synergistic transactivation of the LPL promoter by LXRα/RXRα and FOXA2. Physical and functional interactions between LXRα and FOXA2 were established in vitro and ex vivo. In summary, the present study revealed a novel mechanism of human LPL gene induction by oxysterols in the liver with is based on physical and functional interactions between transcription factors LXRα and FOXA2. This mechanism, which may not be restricted to the LPL gene, is critically important for a better understanding of the regulation of cholesterol and triglyceride metabolism in the liver under healthy or pathological states.
Collapse
Affiliation(s)
- Maria Kanaki
- Laboratory of Biochemistry, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 71003, Greece
| | - Ioanna Tiniakou
- Laboratory of Biochemistry, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 71003, Greece
| | - Efstathia Thymiakou
- Laboratory of Biochemistry, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 71003, Greece
| | - Dimitris Kardassis
- Laboratory of Biochemistry, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 71003, Greece,.
| |
Collapse
|
35
|
Kanaki M, Kardassis D. Regulation of the human lipoprotein lipase gene by the forkhead box transcription factor FOXA2/HNF-3β in hepatic cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:327-336. [DOI: 10.1016/j.bbagrm.2017.01.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/13/2017] [Accepted: 01/13/2017] [Indexed: 12/11/2022]
|
36
|
Akbar S, Pinçon A, Lanhers MC, Claudepierre T, Corbier C, Gregory-Pauron L, Malaplate-Armand C, Visvikis A, Oster T, Yen FT. Expression profile of hepatic genes related to lipid homeostasis in LSR heterozygous mice contributes to their increased response to high-fat diet. Physiol Genomics 2016; 48:928-935. [PMID: 27789735 DOI: 10.1152/physiolgenomics.00077.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/19/2016] [Indexed: 12/30/2022] Open
Abstract
Perturbations of lipid homeostasis manifest as dyslipidemias and obesity, which are significant risk factors for atherosclerosis and diabetes. Lipoprotein receptors in the liver are key players in the regulation of lipid homeostasis, among which the hepatic lipolysis stimulated lipoprotein receptor, LSR, was recently shown to play an important role in the removal of lipoproteins from the circulation during the postprandial phase. Since heterozygous LSR+/- mice demonstrate moderate dyslipidemia and develop higher body weight gain in response to high-fat diet compared with littermate LSR+/+ controls, we questioned if LSR heterozygosity could affect genes related to hepatic lipid metabolism. A target-specific qPCR array for 84 genes related to lipid metabolism was performed on mRNA isolated from livers of 6 mo old female LSR+/- mice and LSR+/+ littermates following a 6 wk period on a standard (STD) or high-fat diet (60% kcal, HFD). Of the 84 genes studied, 32 were significantly downregulated in STD-LSR+/- mice compared with STD-LSR+/+, a majority of which were PPARα target genes involved in lipid metabolism and transport, and insulin and adipokine-signaling pathways. Of these 32 genes, 80% were also modified in HFD-LSR+/+, suggesting that STD-LSR+/- mice demonstrated a predisposition towards a "high-fat"-like profile, which could reflect dysregulation of liver lipid homeostasis. Since similar profiles of genes were affected by either LSR heterozygosity or by high-fat diet, this would suggest that LSR is a key receptor in regulating hepatic lipid homeostasis, and whose downregulation combined with a Western-type diet may increase predisposition to diet-induced obesity.
Collapse
Affiliation(s)
- Samina Akbar
- EA3998 INRA USC 0340 UR AFPA, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Anthony Pinçon
- EA3998 INRA USC 0340 UR AFPA, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Marie-Claire Lanhers
- EA3998 INRA USC 0340 UR AFPA, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Thomas Claudepierre
- EA3998 INRA USC 0340 UR AFPA, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Catherine Corbier
- EA3998 INRA USC 0340 UR AFPA, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Lynn Gregory-Pauron
- EA3998 INRA USC 0340 UR AFPA, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | | | - Athanase Visvikis
- UMR 7365 CNRS IMOPA, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Thierry Oster
- EA3998 INRA USC 0340 UR AFPA, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Frances T Yen
- EA3998 INRA USC 0340 UR AFPA, Université de Lorraine, Vandœuvre-lès-Nancy, France; .,INSERM, Nancy, France; and
| |
Collapse
|