1
|
Kaminskas Fernandes Isern C, Chen Y, Touboul R, Frank B, Hu S, Chang CL. Exploring the Synergistic Action of Medium-Chain Triglycerides and Omega-3 Fatty Acids to Enhance Cellular Uptake and Anti-Inflammatory Responses. Nutrients 2025; 17:1889. [PMID: 40507158 PMCID: PMC12158061 DOI: 10.3390/nu17111889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2025] [Revised: 05/21/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
Objectives: Omega-3 fatty acids (n-3 FA) exhibit pro-healing and anti-inflammatory properties. Injectable lipid emulsions containing n-3 FA are being explored for the treatment of acute adverse conditions. Our previous studies demonstrated that a triglyceride (TG)-rich emulsion (TGRP) containing medium-chain TG (MCT) and n-3 TG (8:2 ratio) is rapidly cleared from the blood and efficiently taken up by organs. This study systematically examined the impact of varying MCT:n-3 ratios on cellular uptake and metabolic function in inflammatory processes. Methods and results: We measured the uptake of radio-labeled TGRP, comprising pure MCT, n-3, or mixed at selected ratios (8:2, 6:4, 2:8), both in vitro and in vivo. Murine macrophages with MCT:n-3 (6:4 or 2:8) had a 2-fold higher TG uptake. IV-injected mixed TGRP also enhanced blood clearance and organ uptake. n-3 TGRP reduced LPS-induced pro-inflammatory cytokines (TNF-α, IL-1, IL-6) in a dose-dependent manner. The 8:2 ratio enhanced mitochondrial respiration and glycolytic capacity in macrophages. Pro-inflammatory lipids decreased with MCT:n-3 (2:8) and pure n-3 TGRP. Bolus injections of n-3 TGRP with MCT lowered LPS-induced IL-6 in plasma and tissues. Conclusions: MCT and n-3 FA support metabolic activity and exhibit anti-inflammatory effects, suggesting that optimizing their ratio may enhance the therapeutic effects of emulsions for inflammatory conditions.
Collapse
Affiliation(s)
| | - Yao Chen
- Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Roni Touboul
- Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Benjamin Frank
- Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Shuchen Hu
- Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Chuchun L. Chang
- Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
2
|
Xu J, Peng H, Kuang R, Han Z, Zhou H, Hu M, Guo Y, Xu Z, Wang D, Ma R, Takao D, Zhu M, Li F, Zhao Y. Single-cell transcriptome reveals three types of adipocytes associated with intramuscular fat content in pigs. Genomics 2025; 117:110998. [PMID: 39855485 DOI: 10.1016/j.ygeno.2025.110998] [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: 10/29/2024] [Revised: 12/27/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
Intramuscular fat is an essential component of muscle tissue, and understanding its contribution to skeletal muscle fat infiltration and meat quality, together with the underlying genetic mechanisms, is a major topic in pig husbandry. However, the composition of cell types and gene expression profiles essential for this purpose remain largely unexplored. Here, we performed single-cell transcriptome analysis on muscle tissue from adult pigs and identified 15 cell types, including three previously uncharacterized types of adipocytes: Adipocyte 1, Adipocyte 2, and Aregs. Phenotypic analysis showed their proportions correlated closely with intramuscular fat content. Based on integrated analysis of ATAC-seq with RNA-seq data, Adipocyte 1 and Aregs have gene expression profiles and transcription factor (TF) motif enrichment typical of adipocytes. On the other hand, myogenic TF motifs were enriched in marker gene promoters in Adipocyte 2, suggesting that these cells originate from muscle cells. Moreover, the marker gene promoters and lineage-specific TF expression in these three adipocyte types were conserved between pigs and humans. These findings provide deep insights towards understanding the complexity of mammalian intramuscular adipocyte types and the gene regulation underlying their organization and function.
Collapse
Affiliation(s)
- Jing Xu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology of Huazhong Agricultural University, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
| | - Hao Peng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology of Huazhong Agricultural University, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
| | - Renzhuo Kuang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology of Huazhong Agricultural University, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
| | - Zheyu Han
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology of Huazhong Agricultural University, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
| | - Honghong Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology of Huazhong Agricultural University, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
| | - Mingyang Hu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology of Huazhong Agricultural University, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
| | - YaPing Guo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology of Huazhong Agricultural University, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
| | - Zhixiang Xu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology of Huazhong Agricultural University, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
| | - Daoyuan Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology of Huazhong Agricultural University, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
| | - Ruixian Ma
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology of Huazhong Agricultural University, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
| | - Daisuke Takao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology of Huazhong Agricultural University, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China; Hubei Hongshan Laboratory, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
| | - Mengjin Zhu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology of Huazhong Agricultural University, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
| | - Fenge Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology of Huazhong Agricultural University, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China.
| | - Yunxia Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology of Huazhong Agricultural University, 1 Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China; Yazhouwan National Laboratory, 8 huanjin Road, Yazhou District, Sanya, City, Hainan Province 572024, China.
| |
Collapse
|
3
|
Huang Y, Chen L, Li L, Qi Y, Tong H, Wu H, Xu J, Leng L, Cheema S, Sun G, Xia Z, McGuire J, Rodrigues B, Young LH, Bucala R, Qi D. Downregulation of adipose LPL by PAR2 contributes to the development of hypertriglyceridemia. JCI Insight 2024; 9:e173240. [PMID: 38973609 PMCID: PMC11383372 DOI: 10.1172/jci.insight.173240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 05/22/2024] [Indexed: 07/09/2024] Open
Abstract
Lipoprotein lipase (LPL) hydrolyzes circulating triglycerides (TGs), releasing fatty acids (FA) and promoting lipid storage in white adipose tissue (WAT). However, the mechanisms regulating adipose LPL and its relationship with the development of hypertriglyceridemia are largely unknown. WAT from obese humans exhibited high PAR2 expression, which was inversely correlated with the LPL gene. Decreased LPL expression was also inversely correlated with elevated plasma TG levels, suggesting that adipose PAR2 might regulate hypertriglyceridemia by downregulating LPL. In mice, aging and high palmitic acid diet (PD) increased PAR2 expression in WAT, which was associated with a high level of macrophage migration inhibitory factor (MIF). MIF downregulated LPL expression and activity in adipocytes by binding with CXCR2/4 receptors and inhibiting Akt phosphorylation. In a MIF overexpression model, high-circulating MIF levels suppressed adipose LPL, and this suppression was associated with increased plasma TGs but not FA. Following PD feeding, adipose LPL expression and activity were significantly reduced, and this reduction was reversed in Par2-/- mice. Recombinant MIF infusion restored high plasma MIF levels in Par2-/- mice, and the levels decreased LPL and attenuated adipocyte lipid storage, leading to hypertriglyceridemia. These data collectively suggest that downregulation of adipose LPL by PAR2/MIF may contribute to the development of hypertriglyceridemia.
Collapse
Affiliation(s)
- Yiheng Huang
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Liujun Chen
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lisha Li
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yadan Qi
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Haibin Tong
- College of Life and Environment Sciences, Wenzhou University, Wenzhou, Zhejiang, China
| | - Hong Wu
- Institute of Cardiovascular Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jinjie Xu
- Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Lin Leng
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Guang Sun
- Faculty of Medicine, Memorial University, St. John’s, Newfoundland, Canada
| | - Zhengyuan Xia
- Guangdong Medical University, Zhanjiang, Guangdong, China
| | - John McGuire
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Brian Rodrigues
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lawrence H. Young
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dake Qi
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Faculty of Medicine, Memorial University, St. John’s, Newfoundland, Canada
| |
Collapse
|
4
|
Raj RR, Lofquist S, Lee MJ. Remodeling of Adipose Tissues by Fatty Acids: Mechanistic Update on Browning and Thermogenesis by n-3 Polyunsaturated Fatty Acids. Pharm Res 2023; 40:467-480. [PMID: 36050546 DOI: 10.1007/s11095-022-03377-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022]
Abstract
Enhancing thermogenesis by increasing the amount and activity of brown and brite adipocytes is a potential therapeutic target for obesity and its associated diseases. Diet plays important roles in energy metabolism and a myriad of dietary components including lipids are known to regulate thermogenesis through recruitment and activation of brown and brite adipocytes. Depending on types of fatty acids (FAs), the major constituent in lipids, their health benefits differ. Long-chain polyunsaturated FAs (PUFAs), especially n-3 PUFAs remodel adipose tissues in a healthier manner with reduced inflammation and enhanced thermogenesis, while saturated FAs exhibit contrasting effects. Lipid mediators derived from FAs act as autocrine/paracrine as well as endocrine factors to regulate thermogenesis. We discuss lipid mediators that may contribute to the differential effects of FAs on adipose tissue remodeling and hence, cardiometabolic diseases. We also discuss current understanding of molecular and cellular mechanisms through which n-3 PUFAs enhance thermogenesis. Elucidating molecular details of beneficial effects of n-3 PUFAs on thermogenesis is expected to provide information that can be used for development of novel therapeutics for obesity and its associated diseases.
Collapse
Affiliation(s)
- Radha Raman Raj
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, 1955 East West Road, Honolulu, HI, 98622, USA
| | - Sydney Lofquist
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, 1955 East West Road, Honolulu, HI, 98622, USA
| | - Mi-Jeong Lee
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, 1955 East West Road, Honolulu, HI, 98622, USA.
| |
Collapse
|
5
|
Liu X, He M, Gan X, Yang Y, Hou Q, Hu R. The Effects of Six Weeks of Fasted Aerobic Exercise on Body Shape and Blood Biochemical Index in Overweight and Obese Young Adult Males. J Exerc Sci Fit 2023; 21:95-103. [PMID: 36447628 PMCID: PMC9674552 DOI: 10.1016/j.jesf.2022.11.003] [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: 08/30/2021] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND/OBJECTIVE The effects of fasted aerobic exercise on body composition and whether it causes adverse effects remain controversial. This study was to compare the effects of fasted and non-fasted aerobic exercise on body shape and blood biochemical indexes in overweight and obese young adult males, and observe whether FAE triggers adverse reactions. METHODS Thirty overweight and obese young adult males were randomly divided into fasted aerobic exercise (FAE) group, non-fasted aerobic exercise (NFAE) group, and control group. They were subjected to indoor treadmill intervention five days a week combined with diet control for six weeks. The FAE group had breakfast 0.5 h after exercise, and the NFAE group exercised 1 h after breakfast. Both groups filled out adverse reaction questionnaires during exercise, and the control group did not have any intervention. Height, weight, body mass index (BMI), and body fat percentage of the three groups of subjects before and after the experiment were measured by the GAIA KIKO bio-resistance antibody composition analyzer in Korea; waist circumference (WC), hip circumference (HC), waist-to-hip ratio (WHR), and waist-to-height ratio (WHtR) were measured by the tape measure method; fasting plasma glucose (FPG), fasting insulin (FINs), total cholesterol (TC), triglyceride (TG), high density lipoprotein cholesterol (HDL-C), low density lipoprotein cholesterol (LDL-C), very low density lipoprotein (VLDL), and HDL-C/LDL-C were measured by Roche C8000 automatic biochemical analysis instrument. RESULTS Weight, BMI, body fat percentage, WC, HC, WHR, WHtR, TG, TC, LDL-C and VLDL decreased very significantly (P < 0.01) in the FAE and NFAE groups after the 6-week experiment. The decrease in FINS was significant in the FAE group (P < 0.05) and the decrease in HDL-C was very significant in the NFAE group (P < 0.01). There was no significant difference in the frequency of adverse reactions between two groups (P > 0.05). CONCLUSION Six-week FAE and NFAE significantly improved body shape in overweight and obese young adult males, while FAE significantly reduced fasting insulin levels and increased tissue cell sensitivity to insulin. And compared to NFAE, 30 min of FAE in the morning did not increase adverse effects.
Collapse
Affiliation(s)
- Xiaolong Liu
- Sports Medicine Laboratory, Guangxi Normal University, Guilin, 541006, Guangxi, China
- Rehabilitation Institute, Guilin Life and Health Career Technical College, Guilin, 541100, Guangxi, China
| | - Mengxiao He
- Key Laboratory of Human Sports Science, Xinjiang Normal University, Urumqi, 830054, Xinjiang, China
- Department of Physical Education and Health, Guilin College, Guilin, 541006, Guangxi, China
| | - Xiaoli Gan
- College of Foreign Studies, Guangxi Normal University, Guilin, 541006, Guangxi, China
| | - Yang Yang
- Department of Sports Science, Shanghai Institute of Physical Education, Shanghai, 200438, China
| | - Qin Hou
- Department of Gastroenterology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Rongbo Hu
- Chair of Building Realization and Robotics, Technical University of Munich, Munich, 80333, Germany
- Corresponding author. Chair of Building Realization and Robotics, Technical University of Munich, Munich, 80333, Germany.
| |
Collapse
|
6
|
Kim SR, Park HJ, Jung UJ. Anti-adiposity and lipid-lowering effects of schisandrol A in diet-induced obese mice. J Food Biochem 2022; 46:e14501. [PMID: 36332134 DOI: 10.1111/jfbc.14501] [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: 05/03/2022] [Revised: 09/20/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
Lignan schisandrol A (SolA) is known to have antioxidant and anti-inflammatory effects. However, the impact of SolA on obesity is poorly understood. To test the hypothesis that SolA has anti-obesity effects, C57BL/6J mice were fed a high-fat diet with or without SolA (0.006%, w/w) for 16 weeks. SolA decreased visceral fat mass (10%) by increasing energy expenditure and upregulating white adipose tissue thermogenic genes mRNA expression. Furthermore, SolA upregulated adipose Lpl mRNA expression and decreased plasma free fatty acid (FFA), triglyceride (TG), apolipoprotein (apo) B, aspartate aminotransferase levels and TG/HDL-cholesterol and apoB/apoA1 ratios as well as hepatic lipid droplets. Increased hepatic β-oxidation and fecal FFA and TG levels were observed in the SolA-supplemented mice, suggesting an association of its lipid-lowering effect with increased hepatic β-oxidation, fecal fat excretion and adipose Lpl. Conclusionally, this study provides evidence on the protective effects of SolA against adiposity, dyslipidemia and nonalcoholic fatty liver disease in obese mice.
Collapse
Affiliation(s)
- Sang Ryong Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Institute of Life Science & Biotechnology, Kyungpook National University, Daegu, South Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, South Korea
| | - Hyo Jin Park
- Department of Food Science and Nutrition, Kyungpook National University, Daegu, South Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan, South Korea
| |
Collapse
|
7
|
Cruciani S, Garroni G, Pala R, Coradduzza D, Cossu ML, Ginesu GC, Capobianco G, Dessole S, Ventura C, Maioli M. Metformin and vitamin D modulate adipose-derived stem cell differentiation towards the beige phenotype. Adipocyte 2022; 11:356-365. [PMID: 35734882 PMCID: PMC9235891 DOI: 10.1080/21623945.2022.2085417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/30/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) represent an ideal stem cell population for regenerative medicine. ADSC adipogenic differentiation is controlled by the activation of a specific transcriptional program, including epigenetic factors and key adipogenic genes. Under certain conditioned media, ADSCs can differentiate into several phenotypes. We previously demonstrated that bioactive molecules could counteract lipid accumulation and regulate adipogenesis, acting on inflammation and vitamin D metabolism. In the present paper, we aimed at evaluating the effect of metformin and vitamin D in targeting ADSC differentiation towards an intermediate phenotype, as beige adipocytes. We exposed ADSCs to different conditioned media and then we evaluated the levels of expression of main markers of adipogenesis, aP2, LPL and ACOT2. We also analysed the gene and protein expression of thermogenic UCP1 protein, and the expression of PARP1 and the beige specific marker TMEM26. Our results showed a novel effect of metformin and vitamin D not only in inhibiting adipogenesis, but also in inducing a specific 'brown-like' phenotype. These findings pave the way for their possible application in the control of de novo lipogenesis useful for the prevention of obesity and its related metabolic disorders.
Collapse
Affiliation(s)
- Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Giuseppe Garroni
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Renzo Pala
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | | | - Maria Laura Cossu
- General Surgery Unit 2 “Clinica Chirurgica” Medical, Surgical and Experimental Sciences Department, University of Sassari, Sassari, Italy
| | - Giorgio Carlo Ginesu
- General Surgery Unit 2 “Clinica Chirurgica” Medical, Surgical and Experimental Sciences Department, University of Sassari, Sassari, Italy
| | - Giampiero Capobianco
- Department of Medical, Surgical and Experimental Sciences, Gynecologic and Obstetric Clinic, University of Sassari, Sassari, Italy
| | - Salvatore Dessole
- Department of Medical, Surgical and Experimental Sciences, Gynecologic and Obstetric Clinic, University of Sassari, Sassari, Italy
| | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Innovation Accelerator, Consiglio Nazionale delle Ricerche, Bologna, Italy
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Center for Developmental Biology and Reprogramming (CEDEBIOR), Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
8
|
Takahashi M, Yamamuro D, Wakabayashi T, Takei A, Takei S, Nagashima S, Okazaki H, Ebihara K, Yagyu H, Takayanagi Y, Onaka T, Goldberg IJ, Ishibashi S. Loss of myeloid lipoprotein lipase exacerbates adipose tissue fibrosis with collagen VI deposition and hyperlipidemia in leptin-deficient obese mice. J Biol Chem 2022; 298:102322. [PMID: 35926714 PMCID: PMC9440390 DOI: 10.1016/j.jbc.2022.102322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 11/20/2022] Open
Abstract
During obesity, tissue macrophages increase in number and become proinflammatory, thereby contributing to metabolic dysfunction. Lipoprotein lipase (LPL), which hydrolyzes triglyceride in lipoproteins, is secreted by macrophages. However, the role of macrophage-derived LPL in adipose tissue remodeling and lipoprotein metabolism is largely unknown. To clarify these issues, we crossed leptin-deficient Lepob/ob mice with mice lacking the Lpl gene in myeloid cells (Lplm−/m−) to generate Lplm−/m−;Lepob/ob mice. We found the weight of perigonadal white adipose tissue (WAT) was increased in Lplm−/m−;Lepob/ob mice compared with Lepob/ob mice due to substantial accumulation of both adipose tissue macrophages and collagen that surrounded necrotic adipocytes. In the fibrotic epidydimal WAT of Lplm−/m−;Lepob/ob mice, we observed an increase in collagen VI and high mobility group box 1, while α-smooth muscle cell actin, a marker of myofibroblasts, was almost undetectable, suggesting that the adipocytes were the major source of the collagens. Furthermore, the adipose tissue macrophages from Lplm−/m−;Lepob/ob mice showed increased expression of genes related to fibrosis and inflammation. In addition, we determined Lplm−/m−;Lepob/ob mice were more hypertriglyceridemic than Lepob/ob mice. Lplm−/m−;Lepob/ob mice also showed slower weight gain than Lepob/ob mice, which was primarily due to reduced food intake. In conclusion, we discovered that the loss of myeloid Lpl led to extensive fibrosis of perigonadal WAT and hypertriglyceridemia. In addition to illustrating an important role of macrophage LPL in regulation of circulating triglyceride levels, these data show that macrophage LPL protects against fibrosis in obese adipose tissues.
Collapse
Affiliation(s)
- Manabu Takahashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan.
| | - Daisuke Yamamuro
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Tetsuji Wakabayashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Akihito Takei
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Shoko Takei
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Shuichi Nagashima
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Hiroaki Okazaki
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Ken Ebihara
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Hiroaki Yagyu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Yuki Takayanagi
- Division of Brain and Neurophysiology, Department of Physiology, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Tatsushi Onaka
- Division of Brain and Neurophysiology, Department of Physiology, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Ira J Goldberg
- NYU-Langone Medical Center, 435 East 30(th) Street, SB617, New York, NY, 10016
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan.
| |
Collapse
|
9
|
Khine HEE, Sungthong R, Sritularak B, Prompetchara E, Chaotham C. Untapped Pharmaceutical Potential of 4,5,4'-Trihydroxy-3,3'-dimethoxybibenzyl for Regulating Obesity: A Cell-Based Study with a Focus on Terminal Differentiation in Adipogenesis. JOURNAL OF NATURAL PRODUCTS 2022; 85:1591-1602. [PMID: 35679136 DOI: 10.1021/acs.jnatprod.2c00213] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Obesity and its global prevalence has become a threat to human health, while its pharmacotherapy via the application of natural products is still underdeveloped. Here, we probed how 4,5,4'-trihydroxy-3,3'-dimethoxybibenzyl (TDB) derived from an orchid (Dendrobium ellipsophyllum) could exert its roles on the differentiation and function of murine (3T3-L1) and human (PCS-210-010) pre-adipocytes and offer some implications to modulate obesity. Cytotoxic effects of TDB on adipocytes were 2-fold lower than those detected with pre-adipocytes, and no significant difference was detected in cytotoxic profiles between both cell lineages. TDB in a dose-dependent manner decreased cellular lipid accumulation and enhanced lipolysis of both cell lines assessed at early differentiation and during maturation. Underlining molecular mechanisms proved that TBD paused the cell cycle progression by regulating inducers and inhibitors in mitotic clonal expansion, leading to growth arrest of pre-adipocytes at the G0/G1 phase. The compound also governed adipocyte differentiation by repressing expressions of crucial adipogenic regulators and effectors through deactivating the AKT/GSK-3β signaling pathway and activating the AMPK-ACC pathway. To this end, TDB has shown its pharmaceutical potential for modulating adipocyte development and function, and it would be a promising candidate for further assessments as a therapeutic agent to defeat obesity.
Collapse
Affiliation(s)
- Hnin Ei Ei Khine
- Pharmaceutical Sciences and Technology Graduate Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Rungroch Sungthong
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, U.K
| | - Boonchoo Sritularak
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Eakachai Prompetchara
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chatchai Chaotham
- Pharmaceutical Sciences and Technology Graduate Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
10
|
Huang Y, Zhou JH, Zhang H, Canfran-Duque A, Singh AK, Perry RJ, Shulman GI, Fernandez-Hernando C, Min W. Brown adipose TRX2 deficiency activates mtDNA-NLRP3 to impair thermogenesis and protect against diet-induced insulin resistance. J Clin Invest 2022; 132:148852. [PMID: 35202005 PMCID: PMC9057632 DOI: 10.1172/jci148852] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 02/23/2022] [Indexed: 02/03/2023] Open
Abstract
Brown adipose tissue (BAT), a crucial heat-generating organ, regulates whole-body energy metabolism by mediating thermogenesis. BAT inflammation is implicated in the pathogenesis of mitochondrial dysfunction and impaired thermogenesis. However, the link between BAT inflammation and systematic metabolism remains unclear. Herein, we use mice with BAT deficiency of thioredoxin-2 (TRX2), a protein that scavenges mitochondrial reactive oxygen species (ROS), to evaluate the impact of BAT inflammation on metabolism and thermogenesis and its underlying mechanism. Our results show that BAT-specific TRX2 ablation improves systematic metabolic performance via enhancing lipid uptake, which protects mice from diet-induced obesity, hypertriglyceridemia, and insulin resistance. TRX2 deficiency impairs adaptive thermogenesis by suppressing fatty acid oxidation. Mechanistically, loss of TRX2 induces excessive mitochondrial ROS, mitochondrial integrity disruption, and cytosolic release of mitochondrial DNA, which in turn activate aberrant innate immune responses in BAT, including the cGAS/STING and the NLRP3 inflammasome pathways. We identify NLRP3 as a key converging point, as its inhibition reverses both the thermogenesis defect and the metabolic benefits seen under nutrient overload in BAT-specific Trx2-deficient mice. In conclusion, we identify TRX2 as a critical hub integrating oxidative stress, inflammation, and lipid metabolism in BAT, uncovering an adaptive mechanism underlying the link between BAT inflammation and systematic metabolism.
Collapse
Affiliation(s)
- Yanrui Huang
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology
| | - Jenny H Zhou
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology
| | - Haifeng Zhang
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology
| | - Alberto Canfran-Duque
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Comparative Medicine, and
| | - Abhishek K Singh
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Comparative Medicine, and
| | - Rachel J Perry
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Carlos Fernandez-Hernando
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology.,Interdepartmental Program in Vascular Biology and Therapeutics, Department of Comparative Medicine, and
| | - Wang Min
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology
| |
Collapse
|
11
|
Al-Shammari RT, Al-Serri AE, Barhoush SA, Al-Bustan SA. Identification and Characterization of Variants in Intron 6 of the LPL Gene Locus among a Sample of the Kuwaiti Population. Genes (Basel) 2022; 13:genes13040664. [PMID: 35456470 PMCID: PMC9024856 DOI: 10.3390/genes13040664] [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: 02/16/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 11/22/2022] Open
Abstract
Lipoprotein lipase (LPL) is responsible for the hydrolysis of lipoproteins; hence defective LPL is associated with metabolic disorders. Here, we identify certain intronic insertions and deletions (InDels) and single nucleotide polymorphisms (SNPs) in intron 6 of the LPL gene and investigate their associations with different phenotypic characteristics in a cohort of the general Kuwaiti population. Two specific regions of intron 6 of the LPL gene, which contain InDels, were amplified via Sanger sequencing in 729 subjects. Genotypic and allelic frequencies were estimated, and genetic modeling was used to investigate genetic associations of the identified variants with lipid profile, body mass index (BMI), and risk of coronary heart disease (CHD). A total of 16 variants were identified, including 2 InDels, 2 novel SNPs, and 12 known SNPs. The most common variants observed among the population were rs293, rs274, rs295, and rs294. The rs293 “A” insertion showed a significant positive correlation with elevated LDL levels, while rs295 was significantly associated with increased BMI. The rs274 and rs294 variants showed a protective effect of the minor allele with decreased CHD prevalence. These findings shed light on the possible role of LPL intronic variants on metabolic disorders.
Collapse
Affiliation(s)
- Reem T. Al-Shammari
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City 13060, Kuwait; (R.T.A.-S.); (S.A.B.)
- Kuwait Medical Genetic Center, Ministry of Health, Kuwait City 70051, Kuwait
| | - Ahmad E. Al-Serri
- Human Genetics Unit, Department of Pathology, Faculty of Medicine, Kuwait University, Kuwait City 46304, Kuwait;
| | - Sahar A. Barhoush
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City 13060, Kuwait; (R.T.A.-S.); (S.A.B.)
| | - Suzanne A. Al-Bustan
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City 13060, Kuwait; (R.T.A.-S.); (S.A.B.)
- Correspondence: ; Tel.: +965-2498-7130 (ext. 7863)
| |
Collapse
|
12
|
Xu H, Thomas MJ, Kaul S, Kallinger R, Ouweneel AB, Maruko E, Oussaada SM, Jongejan A, Cense HA, Nieuwdorp M, Serlie MJ, Goldberg IJ, Civelek M, Parks BW, Lusis AJ, Knaack D, Schill RL, May SC, Reho JJ, Grobe JL, Gantner B, Sahoo D, Sorci-Thomas MG. Pcpe2, a Novel Extracellular Matrix Protein, Regulates Adipocyte SR-BI-Mediated High-Density Lipoprotein Uptake. Arterioscler Thromb Vasc Biol 2021; 41:2708-2725. [PMID: 34551590 PMCID: PMC8551036 DOI: 10.1161/atvbaha.121.316615] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 08/24/2021] [Indexed: 01/22/2023]
Abstract
Objective To investigate the role of adipocyte Pcpe2 (procollagen C-endopeptidase enhancer 2) in SR-BI (scavenger receptor class BI)-mediated HDL-C (high-density lipoprotein cholesterol) uptake and contributions to adipose lipid storage. Approach and Results Pcpe2, a glycoprotein devoid of intrinsic proteolytic activity, is believed to participate in extracellular protein-protein interactions, supporting SR-BI- mediated HDL-C uptake. In published studies, Pcpe2 deficiency increased the development of atherosclerosis by reducing SR-BI-mediated HDL-C catabolism, but the biological impact of this deficiency on adipocyte SR-BI-mediated HDL-C uptake is unknown. Differentiated cells from Ldlr-/-/Pcpe2-/- (Pcpe2-/-) mouse adipose tissue showed elevated SR-BI protein levels, but significantly reduced HDL-C uptake compared to Ldlr-/- (control) adipose tissue. SR-BI-mediated HDL-C uptake was restored by preincubation of cells with exogenous Pcpe2. In diet-fed mice lacking Pcpe2, significant reductions in visceral, subcutaneous, and brown adipose tissue mass were observed, despite elevations in plasma triglyceride and cholesterol concentrations. Significant positive correlations exist between adipose mass and Pcpe2 expression in both mice and humans. Conclusions Overall, these findings reveal a novel and unexpected function for Pcpe2 in modulating SR-BI expression and function as it relates to adipose tissue expansion and cholesterol balance in both mice and humans.
Collapse
Affiliation(s)
- Hao Xu
- Department of Medicine, Division of Endocrinology and Molecular Medicine
| | - Michael J. Thomas
- Pharmacology & Toxicology and
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sushma Kaul
- Department of Medicine, Division of Endocrinology and Molecular Medicine
| | | | - Amber B. Ouweneel
- Department of Medicine, Division of Endocrinology and Molecular Medicine
| | - Elisa Maruko
- Department of Medicine, Division of Endocrinology and Molecular Medicine
| | - Sabrina M. Oussaada
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| | - Aldo Jongejan
- Department of Bioinformatics, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| | - Huib A. Cense
- Department of Surgery, Rode Kruis Ziekenhuis, Beverwijk, the Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| | - Mireille J. Serlie
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| | - Ira J. Goldberg
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, New York University Langone School of Medicine, New York, NY
| | - Mete Civelek
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Brian W. Parks
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Aldons J. Lusis
- Department of Medicine, Human Genetics, Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, California
| | - Darcy Knaack
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Rebecca L. Schill
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sarah C. May
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - John J. Reho
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Comprehensive Rodent Metabolic Phenotyping Core
| | - Justin L. Grobe
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Comprehensive Rodent Metabolic Phenotyping Core
- Department of Biomedical Engineering
| | - Benjamin Gantner
- Department of Medicine, Division of Endocrinology and Molecular Medicine
| | - Daisy Sahoo
- Department of Medicine, Division of Endocrinology and Molecular Medicine
- Pharmacology & Toxicology and
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mary G. Sorci-Thomas
- Department of Medicine, Division of Endocrinology and Molecular Medicine
- Pharmacology & Toxicology and
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
13
|
Cabodevilla AG, Tang S, Lee S, Mullick AE, Aleman JO, Hussain MM, Sessa WC, Abumrad NA, Goldberg IJ. Eruptive xanthoma model reveals endothelial cells internalize and metabolize chylomicrons, leading to extravascular triglyceride accumulation. J Clin Invest 2021; 131:145800. [PMID: 34128469 PMCID: PMC8203467 DOI: 10.1172/jci145800] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/28/2021] [Indexed: 12/13/2022] Open
Abstract
Although tissue uptake of fatty acids from chylomicrons is primarily via lipoprotein lipase (LpL) hydrolysis of triglycerides (TGs), studies of patients with genetic LpL deficiency suggest additional pathways deliver dietary lipids to tissues. Despite an intact endothelial cell (EC) barrier, hyperchylomicronemic patients accumulate chylomicron-derived lipids within skin macrophages, leading to the clinical finding eruptive xanthomas. We explored whether an LpL-independent pathway exists for transfer of circulating lipids across the EC barrier. We found that LpL-deficient mice had a marked increase in aortic EC lipid droplets before and after a fat gavage. Cultured ECs internalized chylomicrons, which were hydrolyzed within lysosomes. The products of this hydrolysis fueled lipid droplet biogenesis in ECs and triggered lipid accumulation in cocultured macrophages. EC chylomicron uptake was inhibited by competition with HDL and knockdown of the scavenger receptor-BI (SR-BI). In vivo, SR-BI knockdown reduced TG accumulation in aortic ECs and skin macrophages of LpL-deficient mice. Thus, ECs internalize chylomicrons, metabolize them in lysosomes, and either store or release their lipids. This latter process may allow accumulation of TGs within skin macrophages and illustrates a pathway that might be responsible for creation of eruptive xanthomas.
Collapse
Affiliation(s)
- Ainara G. Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Songtao Tang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Sungwoon Lee
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Jose O. Aleman
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - M. Mahmood Hussain
- Diabetes and Obesity Center, NYU–Long Island School of Medicine, Mineola, New York, USA
| | - William C. Sessa
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nada A. Abumrad
- Nutritional Sciences, Department of Medicine and Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ira J. Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
14
|
Association between the lipoprotein lipase rs1534649 gene polymorphism in intron one with Body Mass Index and High Density Lipoprotein-Cholesterol. Saudi J Biol Sci 2021; 28:4717-4722. [PMID: 34354459 PMCID: PMC8324958 DOI: 10.1016/j.sjbs.2021.04.085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 11/20/2022] Open
Abstract
Lipoprotein lipase (LPL) is an enzyme involved in lipid metabolism and distribution of fatty acids hence its role in the initiation and development of dyslipidemia and adiposity. Single nucleotide polymorphisms (SNPs) across the LPL gene have been associated with dyslipidemia, however, the association with obesity has been limited towards specific populations. This study examined the association between LPL gene polymorphisms with plasma lipid levels and body mass index (BMI) in the Kuwaiti population. We examined a total of 486 adults (303 and 183 females and males respectively) with plasma lipid levels and BMI. DNA samples were genotyped for two LPL gene polymorphisms (rs1534649 and rs28645722) using TaqMan allelic discrimination. The relationship between the genotypes with both plasma lipid levels and BMI were assessed using linear regression using “SNPassoc” package from R statistical software. Using an additive genetic model, linear regression analysis showed the T-allele of rs1534649 to be associated with increased BMI in a dose-dependent trend β = 2.13 (95% CI 1.33–2.94); p = 1.7 × 10−7. In addition, a borderline significance was observed between the T-allele and low levels of high density lipoprotein-cholesterol β = −0.04 (95% CI −0.08, −0.006); p = 0.02. There were no associations between rs28645722 and plasma lipid levels (p > 0.05). However, a trend was observed between the A-allele and increased BMI β = 1.75 (95% CI 0.14–3.35); p = 0.03. Our study shows intron one polymorphism rs1534649 to increase the risk of obesity and dyslipidemia. Our findings warrant further investigation of the mechanism of LPL on the development of obesity along with the role of intron one and its impact on LPL gene activity.
Collapse
Key Words
- BMI
- BMI, body mass index
- CI, confidence intervals
- HWE, Hardy and Weinberg Equilibrium
- Kuwait
- LPL
- LPL, Lipoprotein lipase
- Obesity
- Polymorphism HDL
- SD, standard deviation
- SNPs, Single nucleotide polymorphisms
- TC, total cholesterol, HDL-C, high-density lipoprotein cholesterol, LDL-C, low-density lipoprotein cholesterol
- TG, triglycerides
- VLDL-C, very low-density lipoproteins cholesterol
- glm, general linear model
- β, Beta-coefficient
Collapse
|
15
|
Valladolid-Acebes I, Åvall K, Recio-López P, Moruzzi N, Bryzgalova G, Björnholm M, Krook A, Alonso EF, Ericsson M, Landfors F, Nilsson SK, Berggren PO, Juntti-Berggren L. Lowering apolipoprotein CIII protects against high-fat diet-induced metabolic derangements. SCIENCE ADVANCES 2021; 7:7/11/eabc2931. [PMID: 33712458 PMCID: PMC7954448 DOI: 10.1126/sciadv.abc2931] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 01/27/2021] [Indexed: 02/05/2023]
Abstract
Increased levels of apolipoprotein CIII (apoCIII), a key regulator of lipid metabolism, result in obesity-related metabolic derangements. We investigated mechanistically whether lowering or preventing high-fat diet (HFD)–induced increase in apoCIII protects against the detrimental metabolic consequences. Mice, first fed HFD for 10 weeks and thereafter also given an antisense (ASO) to lower apoCIII, already showed reduced levels of apoCIII and metabolic improvements after 4 weeks, despite maintained obesity. Prolonged ASO treatment reversed the metabolic phenotype due to increased lipase activity and receptor-mediated hepatic uptake of lipids. Fatty acids were transferred to the ketogenic pathway, and ketones were used in brown adipose tissue (BAT). This resulted in no fat accumulation and preserved morphology and function of liver and BAT. If ASO treatment started simultaneously with the HFD, mice remained lean and metabolically healthy. Thus, lowering apoCIII protects against and reverses the HFD-induced metabolic phenotype by promoting physiological insulin sensitivity.
Collapse
Affiliation(s)
- Ismael Valladolid-Acebes
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden
| | - Karin Åvall
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden
| | - Patricia Recio-López
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden
| | - Noah Moruzzi
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden
| | - Galyna Bryzgalova
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden
| | - Marie Björnholm
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, C3, Integrative Physiology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Elena Fauste Alonso
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden.,Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Madelene Ericsson
- Department of Medical Biosciences, Unit of Physiological Chemistry 6M, Umeå University, SE-901 85 Umeå, Sweden
| | - Fredrik Landfors
- Department of Medical Biosciences, Unit of Physiological Chemistry 6M, Umeå University, SE-901 85 Umeå, Sweden
| | - Stefan K Nilsson
- Department of Medical Biosciences, Unit of Physiological Chemistry 6M, Umeå University, SE-901 85 Umeå, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden.,Division of Integrative Bioscience and Biotechnology, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea.,Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637553, Singapore.,Center for Diabetes and Metabolism Research, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, PR China
| | - Lisa Juntti-Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden.
| |
Collapse
|
16
|
Agudelo CW, Samaha G, Garcia-Arcos I. Alveolar lipids in pulmonary disease. A review. Lipids Health Dis 2020; 19:122. [PMID: 32493486 PMCID: PMC7268969 DOI: 10.1186/s12944-020-01278-8] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 05/05/2020] [Indexed: 12/15/2022] Open
Abstract
Lung lipid metabolism participates both in infant and adult pulmonary disease. The lung is composed by multiple cell types with specialized functions and coordinately acting to meet specific physiologic requirements. The alveoli are the niche of the most active lipid metabolic cell in the lung, the type 2 cell (T2C). T2C synthesize surfactant lipids that are an absolute requirement for respiration, including dipalmitoylphosphatidylcholine. After its synthesis and secretion into the alveoli, surfactant is recycled by the T2C or degraded by the alveolar macrophages (AM). Surfactant biosynthesis and recycling is tightly regulated, and dysregulation of this pathway occurs in many pulmonary disease processes. Alveolar lipids can participate in the development of pulmonary disease from their extracellular location in the lumen of the alveoli, and from their intracellular location in T2C or AM. External insults like smoke and pollution can disturb surfactant homeostasis and result in either surfactant insufficiency or accumulation. But disruption of surfactant homeostasis is also observed in many chronic adult diseases, including chronic obstructive pulmonary disease (COPD), and others. Sustained damage to the T2C is one of the postulated causes of idiopathic pulmonary fibrosis (IPF), and surfactant homeostasis is disrupted during fibrotic conditions. Similarly, surfactant homeostasis is impacted during acute respiratory distress syndrome (ARDS) and infections. Bioactive lipids like eicosanoids and sphingolipids also participate in chronic lung disease and in respiratory infections. We review the most recent knowledge on alveolar lipids and their essential metabolic and signaling functions during homeostasis and during some of the most commonly observed pulmonary diseases.
Collapse
Affiliation(s)
- Christina W Agudelo
- Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Ghassan Samaha
- Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Itsaso Garcia-Arcos
- Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, 11203, USA.
| |
Collapse
|
17
|
Mukherjee S, Aseer KR, Yun JW. Roles of Macrophage Colony Stimulating Factor in White and Brown Adipocytes. BIOTECHNOL BIOPROC E 2020. [DOI: 10.1007/s12257-020-0023-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
18
|
Lee SD, Priest C, Bjursell M, Gao J, Arneson DV, Ahn IS, Diamante G, van Veen JE, Massa MG, Calkin AC, Kim J, Andersén H, Rajbhandari P, Porritt M, Carreras A, Ahnmark A, Seeliger F, Maxvall I, Eliasson P, Althage M, Åkerblad P, Lindén D, Cole TA, Lee R, Boyd H, Bohlooly-Y M, Correa SM, Yang X, Tontonoz P, Hong C. IDOL regulates systemic energy balance through control of neuronal VLDLR expression. Nat Metab 2019; 1:1089-1100. [PMID: 32072135 PMCID: PMC7028310 DOI: 10.1038/s42255-019-0127-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Liver X receptors limit cellular lipid uptake by stimulating the transcription of Inducible Degrader of the LDL Receptor (IDOL), an E3 ubiquitin ligase that targets lipoprotein receptors for degradation. The function of IDOL in systemic metabolism is incompletely understood. Here we show that loss of IDOL in mice protects against the development of diet-induced obesity and metabolic dysfunction by altering food intake and thermogenesis. Unexpectedly, analysis of tissue-specific knockout mice revealed that IDOL affects energy balance, not through its actions in peripheral metabolic tissues (liver, adipose, endothelium, intestine, skeletal muscle), but by controlling lipoprotein receptor abundance in neurons. Single-cell RNA sequencing of the hypothalamus demonstrated that IDOL deletion altered gene expression linked to control of metabolism. Finally, we identify VLDLR rather than LDLR as the primary mediator of IDOL effects on energy balance. These studies identify a role for the neuronal IDOL-VLDLR pathway in metabolic homeostasis and diet-induced obesity.
Collapse
Affiliation(s)
- Stephen D Lee
- Department of Pathology and Laboratory Medicine, Department of Biological Chemistry, and Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Christina Priest
- Department of Pathology and Laboratory Medicine, Department of Biological Chemistry, and Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Mikael Bjursell
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jie Gao
- Department of Pathology and Laboratory Medicine, Department of Biological Chemistry, and Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Douglas V Arneson
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - In Sook Ahn
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Graciel Diamante
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - J Edward van Veen
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Megan G Massa
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anna C Calkin
- Department of Pathology and Laboratory Medicine, Department of Biological Chemistry, and Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jason Kim
- Department of Pathology and Laboratory Medicine, Department of Biological Chemistry, and Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Harriet Andersén
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Prashant Rajbhandari
- Department of Pathology and Laboratory Medicine, Department of Biological Chemistry, and Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michelle Porritt
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Alba Carreras
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Andrea Ahnmark
- Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Frank Seeliger
- Pathology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Ingela Maxvall
- Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Pernilla Eliasson
- Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Magnus Althage
- Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Peter Åkerblad
- Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Daniel Lindén
- Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Division of Endocrinology, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tracy A Cole
- Central Nervous System Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA, USA
| | - Richard Lee
- Central Nervous System Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA, USA
| | - Helen Boyd
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca; Cambridge Science Park, Cambridge, UK
| | | | - Stephanie M Correa
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, Department of Biological Chemistry, and Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Cynthia Hong
- Department of Pathology and Laboratory Medicine, Department of Biological Chemistry, and Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
19
|
Chang HR, Josefs T, Scerbo D, Gumaste N, Hu Y, Huggins LA, Barett T, Chiang S, Grossman J, Bagdasarov S, Fisher EA, Goldberg IJ. Role of LpL (Lipoprotein Lipase) in Macrophage Polarization In Vitro and In Vivo. Arterioscler Thromb Vasc Biol 2019; 39:1967-1985. [PMID: 31434492 PMCID: PMC6761022 DOI: 10.1161/atvbaha.119.312389] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Fatty acid uptake and oxidation characterize the metabolism of alternatively activated macrophage polarization in vitro, but the in vivo biology is less clear. We assessed the roles of LpL (lipoprotein lipase)-mediated lipid uptake in macrophage polarization in vitro and in several important tissues in vivo. Approach and Results: We created mice with both global and myeloid-cell specific LpL deficiency. LpL deficiency in the presence of VLDL (very low-density lipoproteins) altered gene expression of bone marrow-derived macrophages and led to reduced lipid uptake but an increase in some anti- and some proinflammatory markers. However, LpL deficiency did not alter lipid accumulation or gene expression in circulating monocytes nor did it change the ratio of Ly6Chigh/Ly6Clow. In adipose tissue, less macrophage lipid accumulation was found with global but not myeloid-specific LpL deficiency. Neither deletion affected the expression of inflammatory genes. Global LpL deficiency also reduced the numbers of elicited peritoneal macrophages. Finally, we assessed gene expression in macrophages from atherosclerotic lesions during regression; LpL deficiency did not affect the polarity of plaque macrophages. CONCLUSIONS The phenotypic changes observed in macrophages upon deletion of Lpl in vitro is not mimicked in tissue macrophages.
Collapse
Affiliation(s)
- Hye Rim Chang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Tatjana Josefs
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Diego Scerbo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Namrata Gumaste
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Yunying Hu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Lesley-Ann Huggins
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Tessa Barett
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York; Division of Vascular Surgery, Department of Surgery, New York University School of Medicine, New York, New York
| | - Stephanie Chiang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Jennifer Grossman
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Svetlana Bagdasarov
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Edward A. Fisher
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Ira J. Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| |
Collapse
|
20
|
Zhang X, Xue C, Lin J, Ferguson JF, Weiner A, Liu W, Han Y, Hinkle C, Li W, Jiang H, Gosai S, Hachet M, Garcia BA, Gregory BD, Soccio RE, Hogenesch JB, Seale P, Li M, Reilly MP. Interrogation of nonconserved human adipose lincRNAs identifies a regulatory role of linc-ADAL in adipocyte metabolism. Sci Transl Med 2019; 10:10/446/eaar5987. [PMID: 29925637 DOI: 10.1126/scitranslmed.aar5987] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 11/27/2017] [Accepted: 05/04/2018] [Indexed: 12/16/2022]
Abstract
Long intergenic noncoding RNAs (lincRNAs) have emerged as important modulators of cellular functions. Most lincRNAs are not conserved among mammals, raising the fundamental question of whether nonconserved adipose-expressed lincRNAs are functional. To address this, we performed deep RNA sequencing of gluteal subcutaneous adipose tissue from 25 healthy humans. We identified 1001 putative lincRNAs expressed in all samples through de novo reconstruction of noncoding transcriptomes and integration with existing lincRNA annotations. One hundred twenty lincRNAs had adipose-enriched expression, and 54 of these exhibited peroxisome proliferator-activated receptor γ (PPARγ) or CCAAT/enhancer binding protein α (C/EBPα) binding at their loci. Most of these adipose-enriched lincRNAs (~85%) were not conserved in mice, yet on average, they showed degrees of expression and binding of PPARγ and C/EBPα similar to those displayed by conserved lincRNAs. Most adipose lincRNAs differentially expressed (n = 53) in patients after bariatric surgery were nonconserved. The most abundant adipose-enriched lincRNA in our subcutaneous adipose data set, linc-ADAL, was nonconserved, up-regulated in adipose depots of obese individuals, and markedly induced during in vitro human adipocyte differentiation. We demonstrated that linc-ADAL interacts with heterogeneous nuclear ribonucleoprotein U (hnRNPU) and insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) at distinct subcellular locations to regulate adipocyte differentiation and lipogenesis.
Collapse
Affiliation(s)
- Xuan Zhang
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Chenyi Xue
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Jennie Lin
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jane F Ferguson
- Division of Cardiovascular Medicine, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Amber Weiner
- Epigenetics Program, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wen Liu
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Yumiao Han
- Epigenetics Program, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christine Hinkle
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wenjun Li
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongfeng Jiang
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing 100029, China
| | - Sager Gosai
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Melanie Hachet
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Benjamin A Garcia
- Epigenetics Program, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brian D Gregory
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Raymond E Soccio
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John B Hogenesch
- Divisions of Human Genetics and Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45267, USA
| | - Patrick Seale
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mingyao Li
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Muredach P Reilly
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA. .,Irving Institute for Clinical and Translational Research, Columbia University, New York, NY 10032, USA
| |
Collapse
|
21
|
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
|
22
|
Hoekstra M, Nahon JE, de Jong LM, Kröner MJ, de Leeuw LR, Van Eck M. Inhibition of PRMT3 activity reduces hepatic steatosis without altering atherosclerosis susceptibility in apoE knockout mice. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1402-1409. [PMID: 30776415 DOI: 10.1016/j.bbadis.2019.02.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 02/07/2019] [Accepted: 02/12/2019] [Indexed: 12/22/2022]
Abstract
The nuclear receptor liver X receptor (LXR) impacts on cholesterol metabolism as well as hepatic lipogenesis via transcriptional regulation. It is proposed that inhibition of the protein arginine methyltransferase 3 (PRMT3) uncouples these two transcriptional pathways in vivo by acting as a specific lipogenic coactivator of LXR. Here we validated the hypothesis that treatment with the allosteric PRMT3 inhibitor SGC707 will diminish the hepatic steatosis extent, while leaving global cholesterol metabolism, important in cholesterol-driven pathologies like atherosclerosis, untouched. For this purpose, 12-week old hyperlipidemic apolipoprotein E knockout mice were fed a Western-type diet for six weeks to induce both hepatic steatosis and atherosclerosis. The mice received 3 intraperitoneal injections with SGC707 or solvent control per week. Mice chronically treated with SGC707 developed less severe hepatic steatosis as exemplified by the 51% reduced (P < 0.05) liver triglyceride levels. In contrast, the extent of in vivo macrophage foam cell formation and aortic root atherosclerosis was not affected by SGC707 treatment. Interestingly, SGC707-treated mice gained 94% less body weight (P < 0.05), which was paralleled by changes in white adipose tissue morphology, i.e. reduction in adipocyte size and browning. In conclusion, we have shown that through PRMT3 inhibitor treatment specific functions of LXR involved in respectively the development of fatty liver disease and atherosclerosis can be uncoupled, resulting in an overall diminished hepatic steatosis extent without a negative impact on atherosclerosis susceptibility. As such, our studies highlight that PRMT3 inhibition may constitute a novel therapeutic approach to limit the development of fatty liver disease in humans.
Collapse
Affiliation(s)
- Menno Hoekstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, the Netherlands..
| | - Joya E Nahon
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, the Netherlands
| | - Laura M de Jong
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, the Netherlands
| | - Mara J Kröner
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, the Netherlands
| | - Lidewij R de Leeuw
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, the Netherlands
| | - Miranda Van Eck
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, the Netherlands
| |
Collapse
|
23
|
Liu C, Li L, Guo D, Lv Y, Zheng X, Mo Z, Xie W. Lipoprotein lipase transporter GPIHBP1 and triglyceride-rich lipoprotein metabolism. Clin Chim Acta 2018; 487:33-40. [PMID: 30218660 DOI: 10.1016/j.cca.2018.09.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/11/2018] [Accepted: 09/11/2018] [Indexed: 02/05/2023]
Abstract
Increased plasma triglyceride serves as an independent risk factor for cardiovascular disease (CVD). Lipoprotein lipase (LPL), which hydrolyzes circulating triglyceride, plays a crucial role in normal lipid metabolism and energy balance. Hypertriglyceridemia is possibly caused by gene mutations resulting in LPL dysfunction. There are many factors that both positively and negatively interact with LPL thereby impacting TG lipolysis. Glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 (GPIHBP1), a newly identified factor, appears essential for transporting LPL to the luminal side of the blood vessel and offering a platform for TG hydrolysis. Numerous lines of evidence indicate that GPIHBP1 exerts distinct functions and plays diverse roles in human triglyceride-rich lipoprotein (TRL) metabolism. In this review, we discuss the GPIHBP1 gene, protein, its expression and function and subsequently focus on its regulation and provide critical evidence supporting its role in TRL metabolism. Underlying mechanisms of action are highlighted, additional studies discussed and potential therapeutic targets reviewed.
Collapse
Affiliation(s)
- Chuhao Liu
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, Hunan, China; 2016 Class of Excellent Doctor, University of South China, Hengyang 421001, Hunan, China
| | - Liang Li
- Department of Pathophysiology, University of South China, Hengyang 421001, Hunan, China
| | - Dongming Guo
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, Hunan, China
| | - Yuncheng Lv
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, Hunan, China
| | - XiLong Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary T2N 4N1, Alberta, Canada; Key Laboratory of Molecular Targets & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China
| | - Zhongcheng Mo
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, Hunan, China.
| | - Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, Hunan, China.
| |
Collapse
|
24
|
Larsson M, Allan CM, Heizer PJ, Tu Y, Sandoval NP, Jung RS, Walzem RL, Beigneux AP, Young SG, Fong LG. Impaired thermogenesis and sharp increases in plasma triglyceride levels in GPIHBP1-deficient mice during cold exposure. J Lipid Res 2018; 59:706-713. [PMID: 29449313 PMCID: PMC5880501 DOI: 10.1194/jlr.m083832] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Indexed: 01/11/2023] Open
Abstract
Glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1), an endothelial cell protein, binds LPL in the subendothelial spaces and transports it to the capillary lumen. In Gpihbp1-/- mice, LPL remains stranded in the subendothelial spaces, causing hypertriglyceridemia, but how Gpihbp1-/- mice respond to metabolic stress (e.g., cold exposure) has never been studied. In wild-type mice, cold exposure increases LPL-mediated processing of triglyceride-rich lipoproteins (TRLs) in brown adipose tissue (BAT), providing fuel for thermogenesis and leading to lower plasma triglyceride levels. We suspected that defective TRL processing in Gpihbp1-/- mice might impair thermogenesis and blunt the fall in plasma triglyceride levels. Indeed, Gpihbp1-/- mice exhibited cold intolerance, but the effects on plasma triglyceride levels were paradoxical. Rather than falling, the plasma triglyceride levels increased sharply (from ∼4,000 to ∼15,000 mg/dl), likely because fatty acid release by peripheral tissues drives hepatic production of TRLs that cannot be processed. We predicted that the sharp increase in plasma triglyceride levels would not occur in Gpihbp1-/-Angptl4-/- mice, where LPL activity is higher and baseline plasma triglyceride levels are lower. Indeed, the plasma triglyceride levels in Gpihbp1-/-Angptl4-/- mice fell during cold exposure. Metabolic studies revealed increased levels of TRL processing in the BAT of Gpihbp1-/-Angptl4-/- mice.
Collapse
Affiliation(s)
- Mikael Larsson
- Departments of Medicine University of California Los Angeles, Los Angeles, CA 90095.
| | - 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
| | - Rosemary L Walzem
- Department of Poultry Science and Faculty of Nutrition, Texas A&M University, College Station, TX 77843
| | - Anne P Beigneux
- 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.
| | - Loren G Fong
- Departments of Medicine University of California Los Angeles, Los Angeles, CA 90095.
| |
Collapse
|
25
|
Aryal B, Singh AK, Zhang X, Varela L, Rotllan N, Goedeke L, Chaube B, Camporez JP, Vatner DF, Horvath TL, Shulman GI, Suárez Y, Fernández-Hernando C. Absence of ANGPTL4 in adipose tissue improves glucose tolerance and attenuates atherogenesis. JCI Insight 2018; 3:97918. [PMID: 29563332 PMCID: PMC5926923 DOI: 10.1172/jci.insight.97918] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 02/14/2018] [Indexed: 12/12/2022] Open
Abstract
Alterations in ectopic lipid deposition and circulating lipids are major risk factors for developing cardiometabolic diseases. Angiopoietin-like protein 4 (ANGPTL4), a protein that inhibits lipoprotein lipase (LPL), controls fatty acid (FA) uptake in adipose and oxidative tissues and regulates circulating triacylglycerol-rich (TAG-rich) lipoproteins. Unfortunately, global depletion of ANGPTL4 results in severe metabolic abnormalities, inflammation, and fibrosis when mice are fed a high-fat diet (HFD), limiting our understanding of the contribution of ANGPTL4 in metabolic disorders. Here, we demonstrate that genetic ablation of ANGPTL4 in adipose tissue (AT) results in enhanced LPL activity, rapid clearance of circulating TAGs, increased AT lipolysis and FA oxidation, and decreased FA synthesis in AT. Most importantly, we found that absence of ANGPTL4 in AT prevents excessive ectopic lipid deposition in the liver and muscle, reducing novel PKC (nPKC) membrane translocation and enhancing insulin signaling. As a result, we observed a remarkable improvement in glucose tolerance in short-term HFD-fed AT-specific Angptl4-KO mice. Finally, lack of ANGPTL4 in AT enhances the clearance of proatherogenic lipoproteins, attenuates inflammation, and reduces atherosclerosis. Together, these findings uncovered an essential role of AT ANGPTL4 in regulating peripheral lipid deposition, influencing whole-body lipid and glucose metabolism and the progression of atherosclerosis.
Collapse
Affiliation(s)
- Binod Aryal
- Vascular Biology and Therapeutics Program
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
| | - Abhishek K. Singh
- Vascular Biology and Therapeutics Program
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
| | - Xinbo Zhang
- Vascular Biology and Therapeutics Program
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
| | - Luis Varela
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
| | - Noemi Rotllan
- Vascular Biology and Therapeutics Program
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
| | | | - Balkrishna Chaube
- Vascular Biology and Therapeutics Program
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
| | | | | | - Tamas L. Horvath
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
| | - Gerald I. Shulman
- Department of Internal Medicine
- Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, and
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
26
|
Chang CL, Garcia-Arcos I, Nyrén R, Olivecrona G, Kim JY, Hu Y, Agrawal RR, Murphy AJ, Goldberg IJ, Deckelbaum RJ. Lipoprotein Lipase Deficiency Impairs Bone Marrow Myelopoiesis and Reduces Circulating Monocyte Levels. Arterioscler Thromb Vasc Biol 2018; 38:509-519. [PMID: 29371243 DOI: 10.1161/atvbaha.117.310607] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 01/10/2018] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Tissue macrophages induce and perpetuate proinflammatory responses, thereby promoting metabolic and cardiovascular disease. Lipoprotein lipase (LpL), the rate-limiting enzyme in blood triglyceride catabolism, is expressed by macrophages in atherosclerotic plaques. We questioned whether LpL, which is also expressed in the bone marrow (BM), affects circulating white blood cells and BM proliferation and modulates macrophage retention within the artery. APPROACH AND RESULTS We characterized blood and tissue leukocytes and inflammatory molecules in transgenic LpL knockout mice rescued from lethal hypertriglyceridemia within 18 hours of life by muscle-specific LpL expression (MCKL0 mice). LpL-deficient mice had ≈40% reduction in blood white blood cell, neutrophils, and total and inflammatory monocytes (Ly6C/Ghi). LpL deficiency also significantly decreased expression of BM macrophage-associated markers (F4/80 and TNF-α [tumor necrosis factor α]), master transcription factors (PU.1 and C/EBPα), and colony-stimulating factors (CSFs) and their receptors, which are required for monocyte and monocyte precursor proliferation and differentiation. As a result, differentiation of macrophages from BM-derived monocyte progenitors and monocytes was decreased in MCKL0 mice. Furthermore, although LpL deficiency was associated with reduced BM uptake and accumulation of triglyceride-rich particles and macrophage CSF-macrophage CSF receptor binding, triglyceride lipolysis products (eg, linoleic acid) stimulated expression of macrophage CSF and macrophage CSF receptor in BM-derived macrophage precursor cells. Arterial macrophage numbers decreased after heparin-mediated LpL cell dissociation and by genetic knockout of arterial LpL. Reconstitution of LpL-expressing BM replenished aortic macrophage density. CONCLUSIONS LpL regulates peripheral leukocyte levels and affects BM monocyte progenitor differentiation and aortic macrophage accumulation.
Collapse
Affiliation(s)
- Chuchun L Chang
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Itsaso Garcia-Arcos
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Rakel Nyrén
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Gunilla Olivecrona
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Ji Young Kim
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Yunying Hu
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Rishi R Agrawal
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Andrew J Murphy
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.)
| | - Ira J Goldberg
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.).
| | - Richard J Deckelbaum
- From Institute of Human Nutrition (C.L.C., J.Y.K., R.R.A., R.J.D.), Division of Preventive Medicine and Nutrition, Department of Medicine (I.G.-A.), Division of Molecular Medicine, Department of Medicine (Y.H., A.J.M., I.J.G.), and Department of Pediatrics (R.J.D.), College of Physicians and Surgeons, Columbia University, New York; Department of Medical Biosciences/Physiological Chemistry, Umeå University, Sweden (R.N., G.O.); Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York (Y.H., I.J.G.); Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.); and Department of Immunology, Monash University, Melbourne, Victoria, Australia (A.J.M.).
| |
Collapse
|
27
|
Abstract
Dietary triglyceride (TG) is the most efficient energy substrate. It is processed and stored at substantially lower metabolic cost than is protein or carbohydrate. In fed animals, circulating TGs are preferentially routed for storage to white adipose tissue (WAT) by angiopoietin-like proteins 3 (A3) and 8 (A8). Here, we show that mice lacking A3 and A8 (A3-/-A8-/- mice) have decreased fat mass and a striking increase in temperature (+1 °C) in the fed (but not fasted) state, without alterations in food intake or physical activity. Subcutaneous WAT (WAT-SQ) from these animals had morphologic and metabolic changes characteristic of beiging. O2 consumption rates (OCRs) and expression of genes involved in both fatty acid synthesis and fatty acid oxidation were increased in WAT-SQ of A3-/-A8-/- mice, but not in their epididymal or brown adipose tissue (BAT). The hyperthermic response to feeding was blocked by maintaining A3-/-A8-/- mice at thermoneutrality or by treating with a β3-adrenergic receptor (AR) antagonist. To determine if sympathetic stimulation was sufficient to increase body temperature in A3-/-A8-/- mice, WT and A3-/-A8-/- animals were maintained at thermoneutrality and then treated with a β3-AR agonist; treatment induced hyperthermia in A3-/-A8-/- , but not WT, mice. Antibody-mediated inactivation of both circulating A3 and A8 induced hyperthermia in WT mice. Together, these data indicate that A3 and A8 are essential for efficient storage of dietary TG and that disruption of these genes increases feeding-induced thermogenesis and energy utilization.
Collapse
|
28
|
Targeting white, brown and perivascular adipose tissue in atherosclerosis development. Eur J Pharmacol 2017; 816:82-92. [DOI: 10.1016/j.ejphar.2017.03.051] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/14/2017] [Accepted: 03/23/2017] [Indexed: 12/31/2022]
|
29
|
Quantification of Bone Fatty Acid Metabolism and Its Regulation by Adipocyte Lipoprotein Lipase. Int J Mol Sci 2017; 18:ijms18061264. [PMID: 28608812 PMCID: PMC5486086 DOI: 10.3390/ijms18061264] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 06/03/2017] [Accepted: 06/05/2017] [Indexed: 12/15/2022] Open
Abstract
Adipocytes are master regulators of energy homeostasis. Although the contributions of classical brown and white adipose tissue (BAT and WAT, respectively) to glucose and fatty acid metabolism are well characterized, the metabolic role of adipocytes in bone marrow remains largely unclear. Here, we quantify bone fatty acid metabolism and its contribution to systemic nutrient handling in mice. Whereas in parts of the skeleton the specific amount of nutrients taken-up from the circulation was lower than in other metabolically active tissues such as BAT or liver, the overall contribution of the skeleton as a whole organ was remarkable, placing it among the top organs involved in systemic glucose as well as fatty acid clearance. We show that there are considerable site-specific variations in bone marrow fatty acid composition throughout the skeleton and that, especially in the tibia, marrow fatty acid profiles resemble classical BAT and WAT. Using a mouse model lacking lipoprotein lipase (LPL), a master regulator of plasma lipid turnover specifically in adipocytes, we show that impaired fatty acid flux leads to reduced amounts of dietary essential fatty acids while there was a profound increase in de novo produced fatty acids in both bone marrow and cortical bone. Notably, these changes in fatty acid profiles were not associated with any gross skeletal phenotype. These results identify LPL as an important regulator of fatty acid transport to skeletal compartments and demonstrate an intricate functional link between systemic and skeletal fatty acid and glucose metabolism.
Collapse
|
30
|
Cui J, Pang J, Lin YJ, Gong H, Wang ZH, Li YX, Li J, Wang Z, Jiang P, Dai DP, Li J, Cai JP, Huang JD, Zhang TM. Adipose-specific deletion of Kif5b exacerbates obesity and insulin resistance in a mouse model of diet-induced obesity. FASEB J 2017; 31:2533-2547. [PMID: 28242773 DOI: 10.1096/fj.201601103r] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 02/07/2017] [Indexed: 12/23/2022]
Abstract
Recent studies have shown that KIF5B (conventional kinesin heavy chain) mediates glucose transporter type 4 translocation and adiponectin secretion in 3T3-L1 adipocytes, suggesting an involvement of KIF5B in the homeostasis of metabolism. However, the in vivo physiologic function of KIF5B in adipose tissue remains to be determined. In this study, adipose-specific Kif5b knockout (F-K5bKO) mice were generated using the Cre-LoxP strategy. F-K5bKO mice had similar body weights to controls fed on a standard chow diet. However, F-K5bKO mice had hyperlipidemia and significant glucose intolerance and insulin resistance. Deletion of Kif5b aggravated the deleterious impact of a high-fat diet (HFD) on body weight gain, hepatosteatosis, glucose tolerance, and systematic insulin sensitivity. These changes were accompanied by impaired insulin signaling, decreased secretion of adiponectin, and increased serum levels of leptin and proinflammatory adipokines. F-K5bKO mice fed on an HFD exhibited lower energy expenditure and thermogenic dysfunction as a result of whitening of brown adipose due to decreased mitochondria biogenesis and down-regulation of key thermogenic gene expression. In conclusion, selective deletion of Kif5b in adipose tissue exacerbates HFD-induced obesity and its associated metabolic disorders, partly through a decrease in energy expenditure, dysregulation of adipokine secretion, and insulin signaling.-Cui, J., Pang, J., Lin, Y.-J., Gong, H., Wang, Z.-H., Li, Y.-X., Li, J., Wang, Z., Jiang, P., Dai, D.-P., Li, J., Cai, J.-P., Huang, J.-D., Zhang, T.-M. Adipose-specific deletion of Kif5b exacerbates obesity and insulin resistance in a mouse model of diet-induced obesity.
Collapse
Affiliation(s)
- Ju Cui
- The Ministry of Health (MOH) Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Jing Pang
- The Ministry of Health (MOH) Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Ya-Jun Lin
- The Ministry of Health (MOH) Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Huan Gong
- The Ministry of Health (MOH) Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Zhen-He Wang
- School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Yun-Xuan Li
- School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Jin Li
- The Ministry of Health (MOH) Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Ping Jiang
- The Ministry of Health (MOH) Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Da-Peng Dai
- The Ministry of Health (MOH) Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Jian Li
- The Ministry of Health (MOH) Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Jian-Ping Cai
- The Ministry of Health (MOH) Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Jian-Dong Huang
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China; .,Shenzhen Institute of Research and Innovation, University of Hong Kong, Hong Kong, China.,The Centre for Synthetic Biology Engineering Research, Shenzhen Institutes of Advanced Technology, Shenzhen, China
| | - Tie-Mei Zhang
- The Ministry of Health (MOH) Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China;
| |
Collapse
|
31
|
Agrawal N, Freitas Corradi P, Gumaste N, Goldberg IJ. Triglyceride Treatment in the Age of Cholesterol Reduction. Prog Cardiovasc Dis 2016; 59:107-118. [PMID: 27544319 PMCID: PMC5364728 DOI: 10.1016/j.pcad.2016.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 08/15/2016] [Indexed: 01/28/2023]
Abstract
Cholesterol reduction has markedly reduced major cardiovascular disease (CVD) events and shown regression of atherosclerosis in some studies. However, CVD has for decades also been associated with increased levels of circulating triglyceride (TG)-rich lipoproteins. Whether this is due to a direct toxic effect of these lipoproteins on arteries or whether this is merely an association is unresolved. More recent genetic analyses have linked genes that modulate TG metabolism with CVD. Moreover, analyses of subgroups of hypertriglyceridemic (HTG) subjects in clinical trials using fibric acid drugs have been interpreted as evidence that TG reduction reduces CVD events. This review will focus on how HTG might cause CVD, whether TG reduction makes a difference, what pathophysiological defects cause HTG, and what options are available for treatment.
Collapse
Affiliation(s)
- Nidhi Agrawal
- Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, NY
| | - Patricia Freitas Corradi
- Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, NY
| | - Namrata Gumaste
- Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, NY
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, NY.
| |
Collapse
|
32
|
Gordts PLSM, Nock R, Son NH, Ramms B, Lew I, Gonzales JC, Thacker BE, Basu D, Lee RG, Mullick AE, Graham MJ, Goldberg IJ, Crooke RM, Witztum JL, Esko JD. ApoC-III inhibits clearance of triglyceride-rich lipoproteins through LDL family receptors. J Clin Invest 2016; 126:2855-66. [PMID: 27400128 DOI: 10.1172/jci86610] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/12/2016] [Indexed: 02/05/2023] Open
Abstract
Hypertriglyceridemia is an independent risk factor for cardiovascular disease, and plasma triglycerides (TGs) correlate strongly with plasma apolipoprotein C-III (ApoC-III) levels. Antisense oligonucleotides (ASOs) for ApoC-III reduce plasma TGs in primates and mice, but the underlying mechanism of action remains controversial. We determined that a murine-specific ApoC-III-targeting ASO reduces fasting TG levels through a mechanism that is dependent on low-density lipoprotein receptors (LDLRs) and LDLR-related protein 1 (LRP1). ApoC-III ASO treatment lowered plasma TGs in mice lacking lipoprotein lipase (LPL), hepatic heparan sulfate proteoglycan (HSPG) receptors, LDLR, or LRP1 and in animals with combined deletion of the genes encoding HSPG receptors and LDLRs or LRP1. However, the ApoC-III ASO did not lower TG levels in mice lacking both LDLR and LRP1. LDLR and LRP1 were also required for ApoC-III ASO-induced reduction of plasma TGs in mice fed a high-fat diet, in postprandial clearance studies, and when ApoC-III-rich or ApoC-III-depleted lipoproteins were injected into mice. ASO reduction of ApoC-III had no effect on VLDL secretion, heparin-induced TG reduction, or uptake of lipids into heart and skeletal muscle. Our data indicate that ApoC-III inhibits turnover of TG-rich lipoproteins primarily through a hepatic clearance mechanism mediated by the LDLR/LRP1 axis.
Collapse
|
33
|
Shao L, Zhou HJ, Zhang H, Qin L, Hwa J, Yun Z, Ji W, Min W. SENP1-mediated NEMO deSUMOylation in adipocytes limits inflammatory responses and type-1 diabetes progression. Nat Commun 2015; 6:8917. [PMID: 26596471 PMCID: PMC4662081 DOI: 10.1038/ncomms9917] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 10/15/2015] [Indexed: 02/08/2023] Open
Abstract
Adipocyte dysfunction correlates with the development of diabetes. Here we show that mice with a adipocyte-specific deletion of the SUMO-specific protease SENP1 gene develop symptoms of type-1 diabetes mellitus (T1DM), including hyperglycaemia and glucose intolerance with mild insulin resistance. Peri-pancreatic adipocytes from SENP1-deficient mice exhibit heightened NF-κB activity and production of proinflammatory cytokines, which induce CCL5 expression in adjacent pancreatic islets and direct cytotoxic effects on pancreatic islets. Mechanistic studies show that SENP1 deletion in adipocytes enhances SUMOylation of the NF-κB essential molecule, NEMO, at lysine 277/309, leading to increased NF-κB activity, cytokine production and pancreatic inflammation. We further show that NF-κB inhibitors could inhibit pre-diabetic cytokine production, β-cell damages and ameliorate the T1DM phenotype in SENP1-deficient mice. Feeding a high-fat diet augments both type-1 and type-2 diabetes phenotypes in SENP1-deficient mice, consistent with the effects on adipocyte-derived NF-κB and cytokine signalling. Our study reveals previously unrecognized mechanism regulating the onset and progression of T1DM associated with adipocyte dysfunction.
Collapse
Affiliation(s)
- Lan Shao
- The First Affiliated Hospital, Center for Translational Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Pathology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, 10 Amistad St, New Haven, Connecticut 06520, USA
| | - Huanjiao Jenny Zhou
- Department of Pathology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, 10 Amistad St, New Haven, Connecticut 06520, USA
| | - Haifeng Zhang
- Department of Pathology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, 10 Amistad St, New Haven, Connecticut 06520, USA
| | - Lingfeng Qin
- Department of Pathology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, 10 Amistad St, New Haven, Connecticut 06520, USA
| | - John Hwa
- Department of Internal Medicine and Section of Cardiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Zhong Yun
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Weidong Ji
- The First Affiliated Hospital, Center for Translational Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wang Min
- The First Affiliated Hospital, Center for Translational Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Pathology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, 10 Amistad St, New Haven, Connecticut 06520, USA
| |
Collapse
|
34
|
Dijk W, Heine M, Vergnes L, Boon MR, Schaart G, Hesselink MKC, Reue K, van Marken Lichtenbelt WD, Olivecrona G, Rensen PCN, Heeren J, Kersten S. ANGPTL4 mediates shuttling of lipid fuel to brown adipose tissue during sustained cold exposure. eLife 2015; 4. [PMID: 26476336 PMCID: PMC4709329 DOI: 10.7554/elife.08428] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 10/16/2015] [Indexed: 12/21/2022] Open
Abstract
Brown adipose tissue (BAT) activation via cold exposure is increasingly scrutinized as a potential approach to ameliorate cardio-metabolic risk. Transition to cold temperatures requires changes in the partitioning of energy substrates, re-routing fatty acids to BAT to fuel non-shivering thermogenesis. However, the mechanisms behind the redistribution of energy substrates to BAT remain largely unknown. Angiopoietin-like 4 (ANGPTL4), a protein that inhibits lipoprotein lipase (LPL) activity, is highly expressed in BAT. Here, we demonstrate that ANGPTL4 is part of a shuttling mechanism that directs fatty acids derived from circulating triglyceride-rich lipoproteins to BAT during cold. Specifically, we show that cold markedly down-regulates ANGPTL4 in BAT, likely via activation of AMPK, enhancing LPL activity and uptake of plasma triglyceride-derived fatty acids. In contrast, cold up-regulates ANGPTL4 in WAT, abolishing a cold-induced increase in LPL activity. Together, our data indicate that ANGPTL4 is an important regulator of plasma lipid partitioning during sustained cold. DOI:http://dx.doi.org/10.7554/eLife.08428.001 The body stores energy in the form of fat molecules. Most of these molecules are stored in white fat cells. Other fat cells, the so-called brown fat cells, consume fats and produce heat to maintain body temperature in cold conditions. The capacity of brown fat cells to consume fats has led researchers to investigate whether brown fat cells might be a key to combat obesity. When an organism is cold, fat is shuttled to the brown fat cells. An enzyme called lipoprotein lipase is involved in a process that allows these fat molecules to be taken up by brown fat cells. However, it was not clear exactly how this process works. A protein called Angiopoietin-like 4 (ANGPTL4) inhibits the activity of lipoprotein lipase in white fat cells and is also found at high levels in brown fat cells. Here, Dijk et al. used genetic and biochemical approaches to study the role of ANGPTL4 in the fat cells of mice. The experiments show that when mice are exposed to cold, the levels of ANGPTL4 decrease in the brown fat cells. This allows the activity of lipoprotein lipase to increase so that these cells are able to take up more fat molecules. However, the opposite happens in white fat cells during cold exposure. The levels of ANGPTL4 increase, which decreases the activity of lipoprotein lipase in white fat cells to allow fat molecules to be shuttled specifically to the brown fat cells. Further experiments suggest that the opposite regulation of ANGPTL4 in brown and white fat cells could be due to a protein called AMPK. This protein is found at higher levels in brown fat cells than in white fat cells and is produced by brown fat cells during cold exposure. Taken together, Dijk et al. show that organs and cells work together to ensure that fat molecules are appropriately distributed to cells in need of energy, such as to brown fat cells during cold. How these findings could be used to stimulate fat consumption by brown fat cells in humans remains open for further investigation. DOI:http://dx.doi.org/10.7554/eLife.08428.002
Collapse
Affiliation(s)
- Wieneke Dijk
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laurent Vergnes
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, United States
| | - Mariëtte R Boon
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden Univeristy Medical Center, Leiden, The Netherlands
| | - Gert Schaart
- Department of Human Movement Sciences, Maastricht University, Maastricht, The Netherlands
| | - Matthijs K C Hesselink
- Department of Human Movement Sciences, Maastricht University, Maastricht, The Netherlands
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, United States
| | | | - Gunilla Olivecrona
- Department of Medical Biosciences/Physiological Chemistry, Umeå University, Umeå, Sweden
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden Univeristy Medical Center, Leiden, The Netherlands
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sander Kersten
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
35
|
Jang H, Bhasin S, Guarneri T, Serra C, Schneider M, Lee MJ, Guo W, Fried SK, Pencina K, Jasuja R. The Effects of a Single Developmentally Entrained Pulse of Testosterone in Female Neonatal Mice on Reproductive and Metabolic Functions in Adult Life. Endocrinology 2015; 156:3737-46. [PMID: 26132920 PMCID: PMC4588815 DOI: 10.1210/en.2015-1117] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Early postnatal exposures to sex steroids have been well recognized to modulate predisposition to diseases of adulthood. There is a complex interplay between timing, duration and dose of endocrine exposures through environmental or dietary sources that may alter the sensitivity of target tissues to the exogenous stimuli. In this study, we determined the metabolic and reproductive programming effects of a single developmentally entrained pulse of testosterone (T) given to female mice in early postnatal period. CD-1 female mice pups were injected with either 5 μg of T enanthate (TE) or vehicle (control [CON] group) within 24 hours after birth and followed to adult age. A total of 66% of T-treated mice exhibited irregular cycling, anovulatory phenotype, and significantly higher ovarian weights than vehicle-treated mice. Longitudinal nuclear magnetic resonance measurements revealed that TE group had greater body weight, whole-body lean, and fat mass than the CON group. Adipose tissue cellularity analysis in TE group revealed a trend toward higher size and number than their littermate CONs. The brown adipose tissue of TE mice exhibited white fat infiltration with down-regulation of several markers, including uncoupling protein 1 (UCP-1), cell death-inducing DNA fragmentation factor, α-subunit-like effector A, bone morphogenetic protein 7 as well as brown adipose tissue differentiation-related transcription regulators. T-injected mice were also more insulin resistant than CON mice. These reproductive and metabolic reprogramming effects were not observed in animals exposed to TE at 3 and 6 weeks of age. Collectively, these data suggest that sustained reproductive and metabolic alterations may result in female mice from a transient exposure to T during a narrow postnatal developmental window.
Collapse
Affiliation(s)
- Hyeran Jang
- Research Program in Men's Health: Aging and Metabolism (H.J., S.B., T.G., C.S., W.G., K.P., R.J.), Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Boston Nutrition and Obesity Research Center (M.S., M.-J.L., S.K.F.), Section of Endocrinology, Diabetes and Nutrition, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Shalender Bhasin
- Research Program in Men's Health: Aging and Metabolism (H.J., S.B., T.G., C.S., W.G., K.P., R.J.), Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Boston Nutrition and Obesity Research Center (M.S., M.-J.L., S.K.F.), Section of Endocrinology, Diabetes and Nutrition, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Tyler Guarneri
- Research Program in Men's Health: Aging and Metabolism (H.J., S.B., T.G., C.S., W.G., K.P., R.J.), Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Boston Nutrition and Obesity Research Center (M.S., M.-J.L., S.K.F.), Section of Endocrinology, Diabetes and Nutrition, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Carlo Serra
- Research Program in Men's Health: Aging and Metabolism (H.J., S.B., T.G., C.S., W.G., K.P., R.J.), Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Boston Nutrition and Obesity Research Center (M.S., M.-J.L., S.K.F.), Section of Endocrinology, Diabetes and Nutrition, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Mary Schneider
- Research Program in Men's Health: Aging and Metabolism (H.J., S.B., T.G., C.S., W.G., K.P., R.J.), Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Boston Nutrition and Obesity Research Center (M.S., M.-J.L., S.K.F.), Section of Endocrinology, Diabetes and Nutrition, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Mi-Jeong Lee
- Research Program in Men's Health: Aging and Metabolism (H.J., S.B., T.G., C.S., W.G., K.P., R.J.), Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Boston Nutrition and Obesity Research Center (M.S., M.-J.L., S.K.F.), Section of Endocrinology, Diabetes and Nutrition, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Wen Guo
- Research Program in Men's Health: Aging and Metabolism (H.J., S.B., T.G., C.S., W.G., K.P., R.J.), Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Boston Nutrition and Obesity Research Center (M.S., M.-J.L., S.K.F.), Section of Endocrinology, Diabetes and Nutrition, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Susan K Fried
- Research Program in Men's Health: Aging and Metabolism (H.J., S.B., T.G., C.S., W.G., K.P., R.J.), Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Boston Nutrition and Obesity Research Center (M.S., M.-J.L., S.K.F.), Section of Endocrinology, Diabetes and Nutrition, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Karol Pencina
- Research Program in Men's Health: Aging and Metabolism (H.J., S.B., T.G., C.S., W.G., K.P., R.J.), Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Boston Nutrition and Obesity Research Center (M.S., M.-J.L., S.K.F.), Section of Endocrinology, Diabetes and Nutrition, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Ravi Jasuja
- Research Program in Men's Health: Aging and Metabolism (H.J., S.B., T.G., C.S., W.G., K.P., R.J.), Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Boston Nutrition and Obesity Research Center (M.S., M.-J.L., S.K.F.), Section of Endocrinology, Diabetes and Nutrition, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| |
Collapse
|
36
|
Abstract
Angiopoietin-like protein 3 (ANGPTL3) is a circulating inhibitor of lipoprotein and endothelial lipase whose physiological function has remained obscure. Here we show that ANGPTL3 plays a major role in promoting uptake of circulating very low density lipoprotein-triglycerides (VLDL-TGs) into white adipose tissue (WAT) rather than oxidative tissues (skeletal muscle, heart brown adipose tissue) in the fed state. This conclusion emerged from studies of Angptl3(-/-) mice. Whereas feeding increased VLDL-TG uptake into WAT eightfold in wild-type mice, no increase occurred in fed Angptl3(-/-) animals. Despite the reduction in delivery to and retention of TG in WAT, fat mass was largely preserved by a compensatory increase in de novo lipogenesis in Angptl3(-/-) mice. Glucose uptake into WAT was increased 10-fold in KO mice, and tracer studies revealed increased conversion of glucose to fatty acids in WAT but not liver. It is likely that the increased uptake of glucose into WAT explains the increased insulin sensitivity associated with inactivation of ANGPTL3. The beneficial effects of ANGPTL3 deficiency on both glucose and lipoprotein metabolism make it an attractive therapeutic target.
Collapse
|
37
|
Liu C, Gates KP, Fang L, Amar MJ, Schneider DA, Geng H, Huang W, Kim J, Pattison J, Zhang J, Witztum JL, Remaley AT, Dong PD, Miller YI. Apoc2 loss-of-function zebrafish mutant as a genetic model of hyperlipidemia. Dis Model Mech 2015; 8:989-98. [PMID: 26044956 PMCID: PMC4527288 DOI: 10.1242/dmm.019836] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 05/29/2015] [Indexed: 12/27/2022] Open
Abstract
Apolipoprotein C-II (APOC2) is an obligatory activator of lipoprotein lipase. Human patients with APOC2 deficiency display severe hypertriglyceridemia while consuming a normal diet, often manifesting xanthomas, lipemia retinalis and pancreatitis. Hypertriglyceridemia is also an important risk factor for development of cardiovascular disease. Animal models to study hypertriglyceridemia are limited, with no Apoc2-knockout mouse reported. To develop a genetic model of hypertriglyceridemia, we generated an apoc2 mutant zebrafish characterized by the loss of Apoc2 function. apoc2 mutants show decreased plasma lipase activity and display chylomicronemia and severe hypertriglyceridemia, which closely resemble the phenotype observed in human patients with APOC2 deficiency. The hypertriglyceridemia in apoc2 mutants is rescued by injection of plasma from wild-type zebrafish or by injection of a human APOC2 mimetic peptide. Consistent with a previous report of a transient apoc2 knockdown, apoc2 mutant larvae have a minor delay in yolk consumption and angiogenesis. Furthermore, apoc2 mutants fed a normal diet accumulate lipid and lipid-laden macrophages in the vasculature, which resemble early events in the development of human atherosclerotic lesions. In addition, apoc2 mutant embryos show ectopic overgrowth of pancreas. Taken together, our data suggest that the apoc2 mutant zebrafish is a robust and versatile animal model to study hypertriglyceridemia and the mechanisms involved in the pathogenesis of associated human diseases. Highlighted Article: Apoc2 loss-of-function zebrafish display severe hypertriglyceridemia, which is characteristic of human patients with defective lipoprotein lipase activity.
Collapse
Affiliation(s)
- Chao Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Keith P Gates
- Sanford Children's Health Research Center, Programs in Genetic Disease and Development and Aging, and Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Longhou Fang
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Marcelo J Amar
- Lipoprotein Metabolism Section, Cardiopulmonary Branch, NHLBI, NIH, Bethesda, MD, USA
| | - Dina A Schneider
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Honglian Geng
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Wei Huang
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jungsu Kim
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jennifer Pattison
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jian Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Joseph L Witztum
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Cardiopulmonary Branch, NHLBI, NIH, Bethesda, MD, USA
| | - P Duc Dong
- Sanford Children's Health Research Center, Programs in Genetic Disease and Development and Aging, and Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Yury I Miller
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
38
|
Affiliation(s)
- Sara N Vallerie
- From the Department of Medicine, Division of Metabolism, Endocrinology and Nutrition (S.N.V., K.E.B.), and Department of Pathology (K.E.B.), Diabetes and Obesity Center of Excellence, University of Washington School of Medicine, Seattle, WA
| | - Karin E Bornfeldt
- From the Department of Medicine, Division of Metabolism, Endocrinology and Nutrition (S.N.V., K.E.B.), and Department of Pathology (K.E.B.), Diabetes and Obesity Center of Excellence, University of Washington School of Medicine, Seattle, WA.
| |
Collapse
|
39
|
Mayoral R, Osborn O, McNelis J, Johnson AM, Oh DY, Izquierdo CL, Chung H, Li P, Traves PG, Bandyopadhyay G, Pessentheiner AR, Ofrecio JM, Cook JR, Qiang L, Accili D, Olefsky JM. Adipocyte SIRT1 knockout promotes PPARγ activity, adipogenesis and insulin sensitivity in chronic-HFD and obesity. Mol Metab 2015; 4:378-91. [PMID: 25973386 PMCID: PMC4421024 DOI: 10.1016/j.molmet.2015.02.007] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 02/20/2015] [Accepted: 02/24/2015] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Adipose tissue is the primary site for lipid deposition that protects the organisms in cases of nutrient excess during obesogenic diets. The histone deacetylase Sirtuin 1 (SIRT1) inhibits adipocyte differentiation by targeting the transcription factor peroxisome proliferator activated-receptor gamma (PPARγ). METHODS To assess the specific role of SIRT1 in adipocytes, we generated Sirt1 adipocyte-specific knockout mice (ATKO) driven by aP2 promoter onto C57BL/6 background. Sirt1 (flx/flx) aP2Cre (+) (ATKO) and Sirt1 (flx/flx) aP2Cre (-) (WT) mice were fed high-fat diet for 5 weeks (short-term) or 15 weeks (chronic-term). Metabolic studies were combined with gene expression analysis and phosphorylation/acetylation patterns in adipose tissue. RESULTS On standard chow, ATKO mice exhibit low-grade chronic inflammation in adipose tissue, along with glucose intolerance and insulin resistance compared with control fed mice. On short-term HFD, ATKO mice become more glucose intolerant, hyperinsulinemic, insulin resistant and display increased inflammation. During chronic HFD, WT mice developed a metabolic dysfunction, higher than ATKO mice, and thereby, knockout mice are more glucose tolerant, insulin sensitive and less inflamed relative to control mice. SIRT1 attenuates adipogenesis through PPARγ repressive acetylation and, in the ATKO mice adipocyte PPARγ was hyperacetylated. This high acetylation was associated with a decrease in Ser273-PPARγ phosphorylation. Dephosphorylated PPARγ is constitutively active and results in higher expression of genes associated with increased insulin sensitivity. CONCLUSION Together, these data establish that SIRT1 downregulation in adipose tissue plays a previously unknown role in long-term inflammation resolution mediated by PPARγ activation. Therefore, in the context of obesity, the development of new therapeutics that activate PPARγ by targeting SIRT1 may provide novel approaches to the treatment of T2DM.
Collapse
Affiliation(s)
- Rafael Mayoral
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA ; Networked Biomedical Research Center, Hepatic and Digestive Diseases (CIBERehd), Monforte de Lemos 3-5, ISC-III, 28029 Madrid, Spain
| | - Olivia Osborn
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA
| | - Joanne McNelis
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA
| | - Andrew M Johnson
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA
| | - Da Young Oh
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA
| | - Cristina Llorente Izquierdo
- Division of Gastroenterology, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA
| | - Heekyung Chung
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA
| | - Pingping Li
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA
| | - Paqui G Traves
- Molecular Neurobiology Laboratory, The Salk Institute, La Jolla, CA 92037, USA
| | - Gautam Bandyopadhyay
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA
| | | | - Jachelle M Ofrecio
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA
| | - Joshua R Cook
- Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Li Qiang
- Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Domenico Accili
- Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Jerrold M Olefsky
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA
| |
Collapse
|
40
|
Puthanveetil P, Wan A, Rodrigues B. Lipoprotein lipase and angiopoietin-like 4 – Cardiomyocyte secretory proteins that regulate metabolism during diabetic heart disease. Crit Rev Clin Lab Sci 2015; 52:138-49. [DOI: 10.3109/10408363.2014.997931] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
41
|
Willecke F, Scerbo D, Nagareddy P, Obunike JC, Barrett TJ, Abdillahi ML, Trent CM, Huggins LA, Fisher EA, Drosatos K, Goldberg IJ. Lipolysis, and not hepatic lipogenesis, is the primary modulator of triglyceride levels in streptozotocin-induced diabetic mice. Arterioscler Thromb Vasc Biol 2014; 35:102-10. [PMID: 25395613 DOI: 10.1161/atvbaha.114.304615] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Diabetic hypertriglyceridemia is thought to be primarily driven by increased hepatic de novo lipogenesis. However, experiments in animal models indicated that insulin deficiency should decrease hepatic de novo lipogenesis and reduce plasma triglyceride levels. APPROACH AND RESULTS To address the discrepancy between human data and genetically altered mouse models, we investigated whether insulin-deficient diabetic mice had triglyceride changes that resemble those in diabetic humans. Streptozotocin-induced insulin deficiency increased plasma triglyceride levels in mice. Contrary to the mouse models with impaired hepatic insulin receptor signaling, insulin deficiency did not reduce hepatic triglyceride secretion and de novo lipogenesis-related gene expression. Diabetic mice had a marked decrease in postprandial triglycerides clearance, which was associated with decreased lipoprotein lipase and peroxisome proliferator-activated receptor α mRNA levels in peripheral tissues and decreased lipoprotein lipase activity in skeletal muscle, heart, and brown adipose tissue. Diabetic heterozygous lipoprotein lipase knockout mice had markedly elevated fasting plasma triglyceride levels and prolonged postprandial triglycerides clearance. CONCLUSIONS Insulin deficiency causes hypertriglyceridemia by decreasing peripheral lipolysis and not by an increase in hepatic triglycerides production and secretion.
Collapse
Affiliation(s)
- Florian Willecke
- From the Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Department of Medicine, Columbia University, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Saha Cardiovascular Research Center, University of Kentucky, Lexington (P.N.); Department of Biological Sciences, New York City College of Technology, City University of New York, Brooklyn (J.C.O.); Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York (T.J.B., E.A.F.); and Department of Pharmacology, Temple University School of Medicine, Center for Translational Medicine, Philadelphia, PA (K.D.)
| | - Diego Scerbo
- From the Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Department of Medicine, Columbia University, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Saha Cardiovascular Research Center, University of Kentucky, Lexington (P.N.); Department of Biological Sciences, New York City College of Technology, City University of New York, Brooklyn (J.C.O.); Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York (T.J.B., E.A.F.); and Department of Pharmacology, Temple University School of Medicine, Center for Translational Medicine, Philadelphia, PA (K.D.)
| | - Prabhakara Nagareddy
- From the Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Department of Medicine, Columbia University, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Saha Cardiovascular Research Center, University of Kentucky, Lexington (P.N.); Department of Biological Sciences, New York City College of Technology, City University of New York, Brooklyn (J.C.O.); Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York (T.J.B., E.A.F.); and Department of Pharmacology, Temple University School of Medicine, Center for Translational Medicine, Philadelphia, PA (K.D.)
| | - Joseph C Obunike
- From the Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Department of Medicine, Columbia University, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Saha Cardiovascular Research Center, University of Kentucky, Lexington (P.N.); Department of Biological Sciences, New York City College of Technology, City University of New York, Brooklyn (J.C.O.); Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York (T.J.B., E.A.F.); and Department of Pharmacology, Temple University School of Medicine, Center for Translational Medicine, Philadelphia, PA (K.D.)
| | - Tessa J Barrett
- From the Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Department of Medicine, Columbia University, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Saha Cardiovascular Research Center, University of Kentucky, Lexington (P.N.); Department of Biological Sciences, New York City College of Technology, City University of New York, Brooklyn (J.C.O.); Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York (T.J.B., E.A.F.); and Department of Pharmacology, Temple University School of Medicine, Center for Translational Medicine, Philadelphia, PA (K.D.)
| | - Mariane L Abdillahi
- From the Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Department of Medicine, Columbia University, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Saha Cardiovascular Research Center, University of Kentucky, Lexington (P.N.); Department of Biological Sciences, New York City College of Technology, City University of New York, Brooklyn (J.C.O.); Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York (T.J.B., E.A.F.); and Department of Pharmacology, Temple University School of Medicine, Center for Translational Medicine, Philadelphia, PA (K.D.)
| | - Chad M Trent
- From the Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Department of Medicine, Columbia University, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Saha Cardiovascular Research Center, University of Kentucky, Lexington (P.N.); Department of Biological Sciences, New York City College of Technology, City University of New York, Brooklyn (J.C.O.); Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York (T.J.B., E.A.F.); and Department of Pharmacology, Temple University School of Medicine, Center for Translational Medicine, Philadelphia, PA (K.D.)
| | - Lesley A Huggins
- From the Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Department of Medicine, Columbia University, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Saha Cardiovascular Research Center, University of Kentucky, Lexington (P.N.); Department of Biological Sciences, New York City College of Technology, City University of New York, Brooklyn (J.C.O.); Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York (T.J.B., E.A.F.); and Department of Pharmacology, Temple University School of Medicine, Center for Translational Medicine, Philadelphia, PA (K.D.)
| | - Edward A Fisher
- From the Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Department of Medicine, Columbia University, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Saha Cardiovascular Research Center, University of Kentucky, Lexington (P.N.); Department of Biological Sciences, New York City College of Technology, City University of New York, Brooklyn (J.C.O.); Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York (T.J.B., E.A.F.); and Department of Pharmacology, Temple University School of Medicine, Center for Translational Medicine, Philadelphia, PA (K.D.)
| | - Konstantinos Drosatos
- From the Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Department of Medicine, Columbia University, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Saha Cardiovascular Research Center, University of Kentucky, Lexington (P.N.); Department of Biological Sciences, New York City College of Technology, City University of New York, Brooklyn (J.C.O.); Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York (T.J.B., E.A.F.); and Department of Pharmacology, Temple University School of Medicine, Center for Translational Medicine, Philadelphia, PA (K.D.)
| | - Ira J Goldberg
- From the Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Department of Medicine, Columbia University, New York (F.W., D.S., M.L.A., C.M.T., L.A.H., I.J.G.); Saha Cardiovascular Research Center, University of Kentucky, Lexington (P.N.); Department of Biological Sciences, New York City College of Technology, City University of New York, Brooklyn (J.C.O.); Division of Cardiology and the Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York (T.J.B., E.A.F.); and Department of Pharmacology, Temple University School of Medicine, Center for Translational Medicine, Philadelphia, PA (K.D.).
| |
Collapse
|
42
|
Mottillo EP, Balasubramanian P, Lee YH, Weng C, Kershaw EE, Granneman JG. Coupling of lipolysis and de novo lipogenesis in brown, beige, and white adipose tissues during chronic β3-adrenergic receptor activation. J Lipid Res 2014; 55:2276-86. [PMID: 25193997 DOI: 10.1194/jlr.m050005] [Citation(s) in RCA: 227] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Chronic activation of β3-adrenergic receptors (β3-ARs) expands the catabolic activity of both brown and white adipose tissue by engaging uncoupling protein 1 (UCP1)-dependent and UCP1-independent processes. The present work examined de novo lipogenesis (DNL) and TG/glycerol dynamics in classic brown, subcutaneous "beige," and classic white adipose tissues during sustained β3-AR activation by CL 316,243 (CL) and also addressed the contribution of TG hydrolysis to these dynamics. CL treatment for 7 days dramatically increased DNL and TG turnover similarly in all adipose depots, despite great differences in UCP1 abundance. Increased lipid turnover was accompanied by the simultaneous upregulation of genes involved in FAS, glycerol metabolism, and FA oxidation. Inducible, adipocyte-specific deletion of adipose TG lipase (ATGL), the rate-limiting enzyme for lipolysis, demonstrates that TG hydrolysis is required for CL-induced increases in DNL, TG turnover, and mitochondrial electron transport in all depots. Interestingly, the effect of ATGL deletion on induction of specific genes involved in FA oxidation and synthesis varied among fat depots. Overall, these studies indicate that FAS and FA oxidation are tightly coupled in adipose tissues during chronic adrenergic activation, and this effect critically depends on the activity of adipocyte ATGL.
Collapse
Affiliation(s)
- Emilio P Mottillo
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI
| | - Priya Balasubramanian
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI
| | - Yun-Hee Lee
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI
| | - Changren Weng
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI
| | - Erin E Kershaw
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - James G Granneman
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI
| |
Collapse
|
43
|
Sha H, Sun S, Francisco AB, Ehrhardt N, Xue Z, Liu L, Lawrence P, Mattijssen F, Guber RD, Panhwar MS, Brenna JT, Shi H, Xue B, Kersten S, Bensadoun A, Péterfy M, Long Q, Qi L. The ER-associated degradation adaptor protein Sel1L regulates LPL secretion and lipid metabolism. Cell Metab 2014; 20:458-70. [PMID: 25066055 PMCID: PMC4156539 DOI: 10.1016/j.cmet.2014.06.015] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 03/17/2014] [Accepted: 06/06/2014] [Indexed: 10/25/2022]
Abstract
Sel1L is an essential adaptor protein for the E3 ligase Hrd1 in the endoplasmic reticulum (ER)-associated degradation (ERAD), a universal quality-control system in the cell; but its physiological role remains unclear. Here we show that mice with adipocyte-specific Sel1L deficiency are resistant to diet-induced obesity and exhibit postprandial hypertriglyceridemia. Further analyses reveal that Sel1L is indispensable for the secretion of lipoprotein lipase (LPL), independent of its role in Hrd1-mediated ERAD and ER homeostasis. Sel1L physically interacts with and stabilizes the LPL maturation complex consisting of LPL and lipase maturation factor 1 (LMF1). In the absence of Sel1L, LPL is retained in the ER and forms protein aggregates, which are degraded primarily by autophagy. The Sel1L-mediated control of LPL secretion is also seen in other LPL-expressing cell types including cardiac myocytes and macrophages. Thus, our study reports a role of Sel1L in LPL secretion and systemic lipid metabolism.
Collapse
Affiliation(s)
- Haibo Sha
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Shengyi Sun
- Graduate Program in Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853, USA
| | - Adam B Francisco
- Department of Animal Science, Cornell University, Ithaca, NY 14853, USA
| | - Nicole Ehrhardt
- Department of Biomedical Sciences, Medical Genetics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Zhen Xue
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Lei Liu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Peter Lawrence
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Frits Mattijssen
- Nutrition Metabolism and Genomics Group, Wageningen University, Wageningen 6703HD, the Netherlands
| | - Robert D Guber
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Muhammad S Panhwar
- Weill Cornell Medical College in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - J Thomas Brenna
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Hang Shi
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Sander Kersten
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; Nutrition Metabolism and Genomics Group, Wageningen University, Wageningen 6703HD, the Netherlands
| | - André Bensadoun
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Miklós Péterfy
- Department of Biomedical Sciences, Medical Genetics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Qiaoming Long
- Laboratory Animal Research Center, Medical College of Soochow University, Suzhou, Jiangsu 215006, China
| | - Ling Qi
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; Graduate Program in Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
44
|
Li YH, Liu L. Apolipoprotein E synthesized by adipocyte and apolipoprotein E carried on lipoproteins modulate adipocyte triglyceride content. Lipids Health Dis 2014; 13:136. [PMID: 25148848 PMCID: PMC4156606 DOI: 10.1186/1476-511x-13-136] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 08/18/2014] [Indexed: 01/02/2023] Open
Abstract
Excessive energy storage of adipose tissue makes contribution to the occurrence and progression of obesity, which accompanies with multiple adverse complications, such as metabolic syndrome, cardiovascular diseases. It is well known that apolipoprotein E, as a component of lipoproteins, performs a key role in maintaining plasma lipoproteins homeostasis. Interestingly, apolipoprotein E is highly expressed in adipocyte and has positive relation with body fat mass. Apolipoprotein E knock-out mice show small fat mass compared to wild type mice. Moreover, adipocyte deficiency in apolipoprotein E shows impaired lipoproeteins internalization and triglyceride accumulation. Apolipopreotein E-deficient lipoproteins can not induce preadipocyte to form round full-lipid adipocyte, whereas apolipoprotein E-containing lipoproteins can. This article mainly reviews the modulation of apolipoprotein E synthesized by adipocyte and apolipoprotein E carried on lipoproteins in adipocyte triglyceride content.
Collapse
Affiliation(s)
| | - Ling Liu
- Department of Cardiology, the Second Xiangya Hospital, Central South University, #139 Middle Renmin Road, Changsha, Hunan 410011, PR China.
| |
Collapse
|
45
|
Lee YS, Kim JW, Osborne O, Oh DY, Sasik R, Schenk S, Chen A, Chung H, Murphy A, Watkins SM, Quehenberger O, Johnson RS, Olefsky JM. Increased adipocyte O2 consumption triggers HIF-1α, causing inflammation and insulin resistance in obesity. Cell 2014; 157:1339-1352. [PMID: 24906151 PMCID: PMC4114226 DOI: 10.1016/j.cell.2014.05.012] [Citation(s) in RCA: 438] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 01/30/2014] [Accepted: 05/01/2014] [Indexed: 12/30/2022]
Abstract
Adipose tissue hypoxia and inflammation have been causally implicated in obesity-induced insulin resistance. Here, we report that, early in the course of high-fat diet (HFD) feeding and obesity, adipocyte respiration becomes uncoupled, leading to increased oxygen consumption and a state of relative adipocyte hypoxia. These events are sufficient to trigger HIF-1α induction, setting off the chronic adipose tissue inflammatory response characteristic of obesity. At the molecular level, these events involve saturated fatty acid stimulation of the adenine nucleotide translocase 2 (ANT2), an inner mitochondrial membrane protein, which leads to the uncoupled respiratory state. Genetic or pharmacologic inhibition of either ANT2 or HIF-1α can prevent or reverse these pathophysiologic events, restoring a state of insulin sensitivity and glucose tolerance. These results reveal the sequential series of events in obesity-induced inflammation and insulin resistance.
Collapse
Affiliation(s)
- Yun Sok Lee
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jung-Whan Kim
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Olivia Osborne
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA 92093, USA
| | - Da Young Oh
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA 92093, USA
| | - Roman Sasik
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA 92093, USA
| | - Simon Schenk
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ai Chen
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA 92093, USA
| | - Heekyung Chung
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anne Murphy
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - Oswald Quehenberger
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA 92093, USA
| | - Randall S Johnson
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, 171 77, Sweden; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK.
| | - Jerrold M Olefsky
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
46
|
Goulbourne CN, Gin P, Tatar A, Nobumori C, Hoenger A, Jiang H, Grovenor CRM, Adeyo O, Esko JD, Goldberg IJ, Reue K, Tontonoz P, Bensadoun A, Beigneux AP, Young SG, Fong LG. The GPIHBP1-LPL complex is responsible for the margination of triglyceride-rich lipoproteins in capillaries. Cell Metab 2014; 19:849-60. [PMID: 24726386 PMCID: PMC4143151 DOI: 10.1016/j.cmet.2014.01.017] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 12/06/2013] [Accepted: 01/17/2014] [Indexed: 12/16/2022]
Abstract
Triglyceride-rich lipoproteins (TRLs) undergo lipolysis by lipoprotein lipase (LPL), an enzyme that is transported to the capillary lumen by an endothelial cell protein, GPIHBP1. For LPL-mediated lipolysis to occur, TRLs must bind to the lumen of capillaries. This process is often assumed to involve heparan sulfate proteoglycans (HSPGs), but we suspected that TRL margination might instead require GPIHBP1. Indeed, TRLs marginate along the heart capillaries of wild-type but not Gpihbp1⁻/⁻ mice, as judged by fluorescence microscopy, quantitative assays with infrared-dye-labeled lipoproteins, and EM tomography. Both cell-culture and in vivo studies showed that TRL margination depends on LPL bound to GPIHBP1. Notably, the expression of LPL by endothelial cells in Gpihbp1⁻/⁻ mice did not restore defective TRL margination, implying that the binding of LPL to HSPGs is ineffective in promoting TRL margination. Our studies show that GPIHBP1-bound LPL is the main determinant of TRL margination.
Collapse
Affiliation(s)
- Chris N Goulbourne
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Peter Gin
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Angelica Tatar
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chika Nobumori
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Andreas Hoenger
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Haibo Jiang
- Department of Materials, University of Oxford, Oxford OX13PH, UK
| | | | - Oludotun Adeyo
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ira J Goldberg
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Karen Reue
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Peter Tontonoz
- Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - André Bensadoun
- Division of Nutritional Science, Cornell University, Ithaca, NY 14853, USA
| | - Anne P Beigneux
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stephen G Young
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Loren G Fong
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
47
|
Kersten S. Physiological regulation of lipoprotein lipase. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:919-33. [PMID: 24721265 DOI: 10.1016/j.bbalip.2014.03.013] [Citation(s) in RCA: 376] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 03/27/2014] [Accepted: 03/30/2014] [Indexed: 01/01/2023]
Abstract
The enzyme lipoprotein lipase (LPL), originally identified as the clearing factor lipase, hydrolyzes triglycerides present in the triglyceride-rich lipoproteins VLDL and chylomicrons. LPL is primarily expressed in tissues that oxidize or store fatty acids in large quantities such as the heart, skeletal muscle, brown adipose tissue and white adipose tissue. Upon production by the underlying parenchymal cells, LPL is transported and attached to the capillary endothelium by the protein GPIHBP1. Because LPL is rate limiting for plasma triglyceride clearance and tissue uptake of fatty acids, the activity of LPL is carefully controlled to adjust fatty acid uptake to the requirements of the underlying tissue via multiple mechanisms at the transcriptional and post-translational level. Although various stimuli influence LPL gene transcription, it is now evident that most of the physiological variation in LPL activity, such as during fasting and exercise, appears to be driven via post-translational mechanisms by extracellular proteins. These proteins can be divided into two main groups: the liver-derived apolipoproteins APOC1, APOC2, APOC3, APOA5, and APOE, and the angiopoietin-like proteins ANGPTL3, ANGPTL4 and ANGPTL8, which have a broader expression profile. This review will summarize the available literature on the regulation of LPL activity in various tissues, with an emphasis on the response to diverse physiological stimuli.
Collapse
Affiliation(s)
- Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703HD Wageningen, The Netherlands
| |
Collapse
|
48
|
Syamsunarno MRAA, Iso T, Yamaguchi A, Hanaoka H, Putri M, Obokata M, Sunaga H, Koitabashi N, Matsui H, Maeda K, Endo K, Tsushima Y, Yokoyama T, Kurabayashi M. Fatty acid binding protein 4 and 5 play a crucial role in thermogenesis under the conditions of fasting and cold stress. PLoS One 2014; 9:e90825. [PMID: 24603714 PMCID: PMC3946242 DOI: 10.1371/journal.pone.0090825] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 02/04/2014] [Indexed: 12/30/2022] Open
Abstract
Hypothermia is rapidly induced during cold exposure when thermoregulatory mechanisms, including fatty acid (FA) utilization, are disturbed. FA binding protein 4 (FABP4) and FABP5, which are abundantly expressed in adipose tissues and macrophages, have been identified as key molecules in the pathogenesis of overnutrition-related diseases, such as insulin resistance and atherosclerosis. We have recently shown that FABP4/5 are prominently expressed in capillary endothelial cells in the heart and skeletal muscle and play a crucial role in FA utilization in these tissues. However, the role of FABP4/5 in thermogenesis remains to be determined. In this study, we showed that thermogenesis is severely impaired in mice lacking both FABP4 and FABP5 (DKO mice), as manifested shortly after cold exposure during fasting. In DKO mice, the storage of both triacylglycerol in brown adipose tissue (BAT) and glycogen in skeletal muscle (SkM) was nearly depleted after fasting, and a biodistribution analysis using 125I-BMIPP revealed that non-esterified FAs (NEFAs) are not efficiently taken up by BAT despite the robustly elevated levels of serum NEFAs. In addition to the severe hypoglycemia observed in DKO mice during fasting, cold exposure did not induce the uptake of glucose analogue 18F-FDG by BAT. These findings strongly suggest that DKO mice exhibit pronounced hypothermia after fasting due to the depletion of energy storage in BAT and SkM and the reduced supply of energy substrates to these tissues. In conclusion, FABP4/5 play an indispensable role in thermogenesis in BAT and SkM. Our study underscores the importance of FABP4/5 for overcoming life-threatening environments, such as cold and starvation.
Collapse
Affiliation(s)
| | - Tatsuya Iso
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, Gunma, Japan
- Education and Research Support Center, Gunma University Graduate School of Medicine, Gunma, Japan
- * E-mail:
| | - Aiko Yamaguchi
- Department of Bioimaging Information Analysis, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hirofumi Hanaoka
- Department of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Mirasari Putri
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, Gunma, Japan
- Department of Public Health, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Masaru Obokata
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hiroaki Sunaga
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Gunma, Japan
| | - Norimichi Koitabashi
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hiroki Matsui
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Gunma, Japan
| | - Kazuhisa Maeda
- Department of Complementary and Alternative Medicine, Graduate School of Medicine, Osaka University Hospital, Osaka, Japan
| | - Keigo Endo
- Department of Bioimaging Information Analysis, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Tomoyuki Yokoyama
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Gunma, Japan
| | - Masahiko Kurabayashi
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, Gunma, Japan
- Education and Research Support Center, Gunma University Graduate School of Medicine, Gunma, Japan
| |
Collapse
|
49
|
Larsson M, Vorrsjö E, Talmud P, Lookene A, Olivecrona G. Apolipoproteins C-I and C-III inhibit lipoprotein lipase activity by displacement of the enzyme from lipid droplets. J Biol Chem 2013; 288:33997-34008. [PMID: 24121499 DOI: 10.1074/jbc.m113.495366] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Apolipoproteins (apo) C-I and C-III are known to inhibit lipoprotein lipase (LPL) activity, but the molecular mechanisms for this remain obscure. We present evidence that either apoC-I or apoC-III, when bound to triglyceride-rich lipoproteins, prevent binding of LPL to the lipid/water interface. This results in decreased lipolytic activity of the enzyme. Site-directed mutagenesis revealed that hydrophobic amino acid residues centrally located in the apoC-III molecule are critical for attachment to lipid emulsion particles and consequently inhibition of LPL activity. Triglyceride-rich lipoproteins stabilize LPL and protect the enzyme from inactivating factors such as angiopoietin-like protein 4 (angptl4). The addition of either apoC-I or apoC-III to triglyceride-rich particles severely diminished their protective effect on LPL and rendered the enzyme more susceptible to inactivation by angptl4. These observations were seen using chylomicrons as well as the synthetic lipid emulsion Intralipid. In the presence of the LPL activator protein apoC-II, more of apoC-I or apoC-III was needed for displacement of LPL from the lipid/water interface. In conclusion, we show that apoC-I and apoC-III inhibit lipolysis by displacing LPL from lipid emulsion particles. We also propose a role for these apolipoproteins in the irreversible inactivation of LPL by factors such as angptl4.
Collapse
Affiliation(s)
- Mikael Larsson
- Department of Medical Biosciences/Physiological Chemistry, Umeå University, SE-901 87 Umeå, Sweden
| | - Evelina Vorrsjö
- Department of Medical Biosciences/Physiological Chemistry, Umeå University, SE-901 87 Umeå, Sweden
| | - Philippa Talmud
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, 5 University Street, London WC1E 6JF, United Kingdom
| | - Aivar Lookene
- Department of Chemistry, Tallinn University of Technology, Tallinn 12618, Estonia
| | - Gunilla Olivecrona
- Department of Medical Biosciences/Physiological Chemistry, Umeå University, SE-901 87 Umeå, Sweden.
| |
Collapse
|
50
|
Mice lacking ANGPTL8 (Betatrophin) manifest disrupted triglyceride metabolism without impaired glucose homeostasis. Proc Natl Acad Sci U S A 2013; 110:16109-14. [PMID: 24043787 DOI: 10.1073/pnas.1315292110] [Citation(s) in RCA: 268] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Angiopoietin-like protein (ANGPTL)8 (alternatively called TD26, RIFL, Lipasin, and Betatrophin) is a newly recognized ANGPTL family member that has been implicated in both triglyceride (TG) and glucose metabolism. Hepatic overexpression of ANGPTL8 causes hypertriglyceridemia and increased insulin secretion. Here we examined the effects of inactivating Angptl8 on TG and glucose metabolism in mice. Angptl8 knockout (Angptl8(-/-)) mice gained weight more slowly than wild-type littermates due to a selective reduction in adipose tissue accretion. Plasma levels of TGs of the Angptl8(-/-) mice were similar to wild-type animals in the fasted state but paradoxically decreased after refeeding. The lower TG levels were associated with both a reduction in very low density lipoprotein secretion and an increase in lipoprotein lipase (LPL) activity. Despite the increase in LPL activity, the uptake of very low density lipoprotein-TG is markedly reduced in adipose tissue but preserved in hearts of fed Angptl8(-/-) mice. Taken together, these data indicate that ANGPTL8 plays a key role in the metabolic transition between fasting and refeeding; it is required to direct fatty acids to adipose tissue for storage in the fed state. Finally, glucose and insulin tolerance testing revealed no alterations in glucose homeostasis in mice fed either a chow or high fat diet. Thus, although absence of ANGPTL8 profoundly disrupts TG metabolism, we found no evidence that it is required for maintenance of glucose homeostasis.
Collapse
|