1
|
Cao Y, Araki M, Nakagawa Y, Deisen L, Lundsgaard A, Kanta JM, Holm S, Johann K, Brings Jacobsen JC, Jähnert M, Schürmann A, Kiens B, Clemmensen C, Shimano H, Fritzen AM, Kleinert M. Dietary medium-chain fatty acids reduce hepatic fat accumulation via activation of a CREBH-FGF21 axis. Mol Metab 2024; 87:101991. [PMID: 39019116 PMCID: PMC11327439 DOI: 10.1016/j.molmet.2024.101991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/01/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024] Open
Abstract
OBJECTIVE Dietary medium-chain fatty acids (MCFAs), characterized by chain lengths of 8-12 carbon atoms, have been proposed to have beneficial effects on glucose and lipid metabolism, yet the underlying mechanisms remain elusive. We hypothesized that MCFA intake benefits metabolic health by inducing the release of hormone-like factors. METHODS The effects of chow diet, high-fat diet rich in long-chain fatty acids (LCFA HFD) fed ad libitum or pair-fed to a high-fat diet rich in MCFA (MCFA HFD) on glycemia, hepatic gene expression, circulating fibroblast growth factor 21 (FGF21), and liver fat content in both wildtype and Fgf21 knockout mice were investigated. The impact of a single oral dose of an MCFA-rich oil on circulating FGF21 and hepatic Fgf21 mRNA expression was assessed. In flag-tagged Crebh knockin mice and liver-specific Crebh knockout mice, fed LCFA HFD or MCFA HFD, active hepatic CREBH and hepatic Fgf21 mRNA abundance were determined, respectively. RESULTS MCFA HFD improves glucose tolerance, enhances glucose clearance into brown adipose tissue, and prevents high-fat diet-induced hepatic steatosis in wildtype mice. These benefits are associated with increased liver expression of CREBH target genes (Apoa4 and Apoc2), including Fgf21. Both acute and chronic intake of dietary MCFAs elevate circulating FGF21. Augmented hepatic Fgf21 mRNA following MCFA HFD intake is accompanied by higher levels of active hepatic CREBH; and MCFA-induced hepatic Fgf21 expression is blocked in mice lacking Crebh. Notably, while feeding male and female Fgf21 wildtype mice MCFA HFD results in reduced liver triacylglycerol (TG) levels, this liver TG-lowering effect is blunted in Fgf21 knockout mice fed MCFA HFD. The reduction in liver TG levels observed with MCFA HFD was independent of weight loss. CONCLUSIONS Dietary MCFAs reduce liver fat accumulation via activation of a CREBH-FGF21 signaling axis.
Collapse
Affiliation(s)
- Ye Cao
- Department of Molecular Physiology of Exercise and Nutrition, German Institute of Human Nutrition (DIfE), Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany
| | - Masaya Araki
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Division of Complex Biosystem Research, Department of Research and Development, Institute of Natural Medicine, University of Toyama, Toyama, Toyama 930-0194, Japan
| | - Yoshimi Nakagawa
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Division of Complex Biosystem Research, Department of Research and Development, Institute of Natural Medicine, University of Toyama, Toyama, Toyama 930-0194, Japan
| | - Luisa Deisen
- Department of Molecular Physiology of Exercise and Nutrition, German Institute of Human Nutrition (DIfE), Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany
| | - Annemarie Lundsgaard
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Josephine M Kanta
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Stephanie Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kornelia Johann
- Department of Molecular Physiology of Exercise and Nutrition, German Institute of Human Nutrition (DIfE), Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany
| | - Jens Christian Brings Jacobsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Markus Jähnert
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Department of Experimental Diabetology, German Institute of Human Nutrition (DIfE), 14558 Potsdam, Germany
| | - Annette Schürmann
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Department of Experimental Diabetology, German Institute of Human Nutrition (DIfE), 14558 Potsdam, Germany; Institute of Nutrition Science, University of Potsdam, Nuthetal, Germany; Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus-Senftenberg, The Brandenburg Medical School Theodor Fontane and The University of Potsdam, 14469 Potsdam, Germany
| | - Bente Kiens
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hitoshi Shimano
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Chiyoda-ku, Tokyo 100-0004, Japan.
| | - Andreas M Fritzen
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Maximilian Kleinert
- Department of Molecular Physiology of Exercise and Nutrition, German Institute of Human Nutrition (DIfE), Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; Institute of Nutrition Science, University of Potsdam, Nuthetal, Germany.
| |
Collapse
|
2
|
Bhale AS, Meilhac O, d'Hellencourt CL, Vijayalakshmi MA, Venkataraman K. Cholesterol transport and beyond: Illuminating the versatile functions of HDL apolipoproteins through structural insights and functional implications. Biofactors 2024; 50:922-956. [PMID: 38661230 DOI: 10.1002/biof.2057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
High-density lipoproteins (HDLs) play a vital role in lipid metabolism and cardiovascular health, as they are intricately involved in cholesterol transport and inflammation modulation. The proteome of HDL particles is indeed complex and distinct from other components in the bloodstream. Proteomics studies have identified nearly 285 different proteins associated with HDL; however, this review focuses more on the 15 or so traditionally named "apo" lipoproteins. Important lipid metabolizing enzymes closely working with the apolipoproteins are also discussed. Apolipoproteins stand out for their integral role in HDL stability, structure, function, and metabolism. The unique structure and functions of each apolipoprotein influence important processes such as inflammation regulation and lipid metabolism. These interactions also shape the stability and performance of HDL particles. HDLs apolipoproteins have multifaceted roles beyond cardiovascular diseases (CVDs) and are involved in various physiological processes and disease states. Therefore, a detailed exploration of these apolipoproteins can offer valuable insights into potential diagnostic markers and therapeutic targets. This comprehensive review article aims to provide an in-depth understanding of HDL apolipoproteins, highlighting their distinct structures, functions, and contributions to various physiological processes. Exploiting this knowledge holds great potential for improving HDL function, enhancing cholesterol efflux, and modulating inflammatory processes, ultimately benefiting individuals by limiting the risks associated with CVDs and other inflammation-based pathologies. Understanding the nature of all 15 apolipoproteins expands our knowledge of HDL metabolism, sheds light on their pathological implications, and paves the way for advancements in the diagnosis, prevention, and treatment of lipid and inflammatory-related disorders.
Collapse
Affiliation(s)
- Aishwarya Sudam Bhale
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Olivier Meilhac
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Pierre, France
| | - Christian Lefebvre d'Hellencourt
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Pierre, France
| | | | - Krishnan Venkataraman
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
3
|
Lu XR, Tao Q, Qin Z, Liu XW, Li SH, Bai LX, Ge WB, Liu YX, Li JY, Yang YJ. A combined transcriptomics and proteomics approach to reveal the mechanism of AEE relieving hyperlipidemia in ApoE -/- mice. Biomed Pharmacother 2024; 173:116400. [PMID: 38484560 DOI: 10.1016/j.biopha.2024.116400] [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: 01/08/2024] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
Hyperlipidemia caused by abnormal lipid metabolism has reached epidemic proportions. This phenomenon is also common in companion animals. Previous studies showed that AEE significantly improves abnormal blood lipids in hyperlipidemia rats and mice, but its mechanism is still not clear enough. In this study, the mechanism and potential key pathways of AEE on improving hyperlipidemia in mice were investigated through the transcriptome and proteome study of ApoE-/- mice liver and the verification study on high-fat HepG2 cells. The results showed that AEE significantly decreased the serum TC and LDL-C levels of hyperlipidemia ApoE-/- mice, and significantly increased the enzyme activity of CYP7A1. After AEE intervention, the results of mice liver transcriptome and proteome showed that differential genes and proteins were enriched in lipid metabolism-related pathways. The results of RT-qPCR showed that AEE significantly regulated the expression of genes related to lipid metabolism in mice liver tissue. AEE significantly upregulated the protein expression of CYP7A1 in hyperlipidemia ApoE-/- mice liver tissue. The results in vitro showed that AEE significantly decreased the levels of TC and TG, and improved lipid deposition in high-fat HepG2 cells. AEE significantly increased the expression of CYP7A1 protein in high-fat HepG2 cells. AEE regulates the expression of genes related to lipid metabolism in high-fat HepG2 cells, mainly by FXR-SHP-CYP7A1 and FGF19-TFEB-CYP7A1 pathways. To sum up, AEE can significantly improve the hyperlipidemia status of ApoE-/- mice and the lipid deposition of high-fat HepG2 cells, and its main pathway is probably the bile acid metabolism-related pathway centered on CYP7A1.
Collapse
Affiliation(s)
- Xiao-Rong Lu
- Key Lab of New Animal Drug of Gansu Province,Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Qi Tao
- Key Lab of New Animal Drug of Gansu Province,Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Zhe Qin
- Key Lab of New Animal Drug of Gansu Province,Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Xi-Wang Liu
- Key Lab of New Animal Drug of Gansu Province,Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Shi-Hong Li
- Key Lab of New Animal Drug of Gansu Province,Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Li-Xia Bai
- Key Lab of New Animal Drug of Gansu Province,Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Wen-Bo Ge
- Key Lab of New Animal Drug of Gansu Province,Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Ya-Xian Liu
- Key Lab of New Animal Drug of Gansu Province,Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Jian-Yong Li
- Key Lab of New Animal Drug of Gansu Province,Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China.
| | - Ya-Jun Yang
- Key Lab of New Animal Drug of Gansu Province,Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China.
| |
Collapse
|
4
|
Hong BV, Agus JK, Tang X, Zheng JJ, Romo EZ, Lei S, Zivkovic AM. Precision Nutrition and Cardiovascular Disease Risk Reduction: the Promise of High-Density Lipoproteins. Curr Atheroscler Rep 2023; 25:663-677. [PMID: 37702886 PMCID: PMC10564829 DOI: 10.1007/s11883-023-01148-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2023] [Indexed: 09/14/2023]
Abstract
PURPOSE OF REVIEW Emerging evidence supports the promise of precision nutritional approaches for cardiovascular disease (CVD) prevention. Here, we discuss current findings from precision nutrition trials and studies reporting substantial inter-individual variability in responses to diets and dietary components relevant to CVD outcomes. We highlight examples where early precision nutrition research already points to actionable intervention targets tailored to an individual's biology and lifestyle. Finally, we make the case for high-density lipoproteins (HDL) as a compelling next generation target for precision nutrition aimed at CVD prevention. HDL possesses complex structural features including diverse protein components, lipids, size distribution, extensive glycosylation, and interacts with the gut microbiome, all of which influence HDL's anti-inflammatory, antioxidant, and cholesterol efflux properties. Elucidating the nuances of HDL structure and function at an individual level may unlock personalized dietary and lifestyle strategies to optimize HDL-mediated atheroprotection and reduce CVD risk. RECENT FINDINGS Recent human studies have demonstrated that HDL particles are key players in the reduction of CVD risk. Our review highlights the role of HDL and the importance of personalized therapeutic approaches to improve their potential for reducing CVD risk. Factors such as diet, genetics, glycosylation, and gut microbiome interactions can modulate HDL structure and function at the individual level. We emphasize that fractionating HDL into size-based subclasses and measuring particle concentration are necessary to understand HDL biology and for developing the next generation of diagnostics and biomarkers. These discoveries underscore the need to move beyond a one-size-fits-all approach to HDL management. Precision nutrition strategies that account for personalized metabolic, genetic, and lifestyle data hold promise for optimizing HDL therapies and function to mitigate CVD risk more potently. While human studies show HDL play a key role in reducing CVD risk, recent findings indicate that factors such as diet, genetics, glycosylation, and gut microbes modulate HDL function at the individual level, underscoring the need for precision nutrition strategies that account for personalized variability to optimize HDL's potential for mitigating CVD risk.
Collapse
Affiliation(s)
- Brian V Hong
- Department of Nutrition, University of California, Davis, Davis, CA, 95616, USA
| | - Joanne K Agus
- Department of Nutrition, University of California, Davis, Davis, CA, 95616, USA
| | - Xinyu Tang
- Department of Nutrition, University of California, Davis, Davis, CA, 95616, USA
| | - Jack Jingyuan Zheng
- Department of Nutrition, University of California, Davis, Davis, CA, 95616, USA
| | - Eduardo Z Romo
- Department of Nutrition, University of California, Davis, Davis, CA, 95616, USA
| | - Susan Lei
- Department of Nutrition, University of California, Davis, Davis, CA, 95616, USA
| | - Angela M Zivkovic
- Department of Nutrition, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
5
|
Ji J, Zhao X, Huang J, Wu X, Xie F, Li L, Wang T, Mi S. Apolipoprotein A-IV of diabetic-foot patients upregulates tumor necrosis factor α expression in microfluidic arterial models. Exp Biol Med (Maywood) 2023; 248:691-701. [PMID: 36775868 PMCID: PMC10408548 DOI: 10.1177/15353702221147562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/21/2022] [Indexed: 02/14/2023] Open
Abstract
Diabetic peripheral arterial atherosclerosis is one of the important characteristics of diabetic foot syndrome. Apolipoprotein (Apo A-IV) participates in various physiological processes, and animal studies have shown that it has roles of anti-atherosclerosis, prevention of platelet aggregation and thrombosis. Apo A-IV glycosylation is closely related to the occurrence and development of diabetic peripheral atherosclerosis. This study aimed to explore the mechanism of diabetic peripheral arterial lesions caused by glycosylated Apo A-IV. Type 2 diabetes mellitus (T2DM) and T2DM with diabetic foot patients (T2DM-F; n = 45, 30) were enrolled in this study, and individuals without diabetes (n = 35) served as normal controls (NC). In T2DM group, serum Apo A-IV content was higher than those in NC and T2DM-F group, as carboxymethyl lysine (CML) glycosylation of Apo A-IV in mixed serum from T2DM-F group was identified to be more significant than those in two other groups. Within a microfluidic arterial chip model, Apo A-IV from T2DM and T2DM-F group significantly increased transcription and protein levels of tumor necrosis factor alpha (TNF-α) in chip arteries, and CML expression was observed in T2DM-F group, which were associated with increased nuclear receptor subfamily 4 group A member 3 (NR4A3) expression. Recombinant human Apo A-IV could reverse the stimulating effect of serum Apo A-IV from T2DM-F group on TNF-α expression, and NR4A3 blocking peptide downregulated TNF-α expression by inhibiting NR4A3 expression. In the chip arteries, Apo A-IV from T2DM and T2DM-F increased TNF-α expression and turn them into a pre-atherosclerotic state, which might be one of the important mechanisms of glycosylated Apo A-IV to induce diabetic peripheral arterial lesions and eventually lead to diabetic foot.
Collapse
Affiliation(s)
- Jun Ji
- Department of Cardiovascular Surgery, University of Chinese Academy of Science Shenzhen Hospital, Shenzhen 518027, China
| | - Xiaoyu Zhao
- Bio-manufacturing Engineering Laboratory, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055 China
| | - Jiajun Huang
- Bio-manufacturing Engineering Laboratory, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055 China
| | - Xuanqin Wu
- Department of Cardiovascular Surgery, University of Chinese Academy of Science Shenzhen Hospital, Shenzhen 518027, China
| | - Fang Xie
- Department of Endocrinology, University of Chinese Academy of Science Shenzhen Hospital, Shenzhen 518027, China
| | - Liang Li
- Department of Cardiovascular Surgery, University of Chinese Academy of Science Shenzhen Hospital, Shenzhen 518027, China
| | - Tao Wang
- Department of Cardiovascular Surgery, University of Chinese Academy of Science Shenzhen Hospital, Shenzhen 518027, China
| | - Shengli Mi
- Bio-manufacturing Engineering Laboratory, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055 China
| |
Collapse
|
6
|
LaRussa Z, Kuo HCN, West K, Shen Z, Wisniewski K, Tso P, Coschigano KT, Lo CC. Increased BAT Thermogenesis in Male Mouse Apolipoprotein A4 Transgenic Mice. Int J Mol Sci 2023; 24:4231. [PMID: 36835642 PMCID: PMC9959433 DOI: 10.3390/ijms24044231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/15/2023] [Accepted: 02/15/2023] [Indexed: 02/23/2023] Open
Abstract
Dietary lipids induce apolipoprotein A4 (APOA4) production and brown adipose tissue (BAT) thermogenesis. Administration of exogenous APOA4 elevates BAT thermogenesis in chow-fed mice, but not high-fat diet (HFD)-fed mice. Chronic feeding of HFD attenuates plasma APOA4 production and BAT thermogenesis in wildtype (WT) mice. In light of these observations, we sought to determine whether steady production of APOA4 could keep BAT thermogenesis elevated, even in the presence of HFD consumption, with an aim toward eventual reduction of body weight, fat mass and plasma lipid levels. Transgenic mice with overexpression of mouse APOA4 in the small intestine (APOA4-Tg mice) produce greater plasma APOA4 than their WT controls, even when fed an atherogenic diet. Thus, we used these mice to investigate the correlation of levels of APOA4 and BAT thermogenesis during HFD consumption. The hypothesis of this study was that overexpression of mouse APOA4 in the small intestine and increased plasma APOA4 production would increase BAT thermogenesis and consequently reduce fat mass and plasma lipids of HFD-fed obese mice. To test this hypothesis, BAT thermogenic proteins, body weight, fat mass, caloric intake, and plasma lipids in male APOA4-Tg mice and WT mice fed either a chow diet or a HFD were measured. When fed a chow diet, APOA4 levels were elevated, plasma triglyceride (TG) levels were reduced, and BAT levels of UCP1 trended upward, while body weight, fat mass, caloric intake, and plasma lipids were comparable between APOA4-Tg and WT mice. After a four-week feeding of HFD, APOA4-Tg mice maintained elevated plasma APOA4 and reduced plasma TG, but UCP1 levels in BAT were significantly elevated in comparison to WT controls; body weight, fat mass and caloric intake were still comparable. After 10-week consumption of HFD, however, while APOA4-Tg mice still exhibited increased plasma APOA4, UCP1 levels and reduced TG levels, a reduction in body weight, fat mass and levels of plasma lipids and leptin were finally observed in comparison to their WT controls and independent of caloric intake. Additionally, APOA4-Tg mice exhibited increased energy expenditure at several time points when measured during the 10-week HFD feeding. Thus, overexpression of APOA4 in the small intestine and maintenance of elevated levels of plasma APOA4 appear to correlate with elevation of UCP1-dependent BAT thermogenesis and subsequent protection against HFD-induced obesity in mice.
Collapse
Affiliation(s)
- Zachary LaRussa
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, and Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - Hsuan-Chih N Kuo
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, and Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - Kathryn West
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, and Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - Zhijun Shen
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, and Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - Kevin Wisniewski
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, and Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45237, USA
| | - Karen T Coschigano
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, and Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - Chunmin C Lo
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, and Diabetes Institute, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
7
|
Integrated Network Pharmacology and Proteomic Analyses of Targets and Mechanisms of Jianpi Tianjing Decoction in Treating Vascular Dementia. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:9021546. [PMID: 36714532 PMCID: PMC9876684 DOI: 10.1155/2023/9021546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/20/2022] [Accepted: 12/29/2022] [Indexed: 01/20/2023]
Abstract
Background Vascular dementia (VD), associated with cerebrovascular injury, is characterized by severe cognitive impairment. Jianpi Tianjing Decoction (JTD) has been widely used to treat VD. However, its molecular targets and mechanisms of action in this treatment remain unclear. This study integrated network pharmacology and proteomics to identify targets and mechanisms of JTD in the treatment of VD and to provide new insights and goals for clinical treatments. Methods Systematic network pharmacology was used to identify active chemical compositions, potential targets, and mechanisms of JTD in VD treatment. Then, a mouse model of VD was induced via transient bilateral common carotid artery occlusion to verify the identified targets and mechanisms of JTD against VD using 4D label-free quantitative proteomics. Results By screening active chemical compositions and potential targets in relevant databases, 187 active chemical compositions and 416 disease-related compound targets were identified. In vivo experiments showed that JTD improved learning and memory in mice. Proteomics also identified 112 differentially expressed proteins in the model and sham groups and the JTD and model groups. Integrating the network pharmacology and proteomics results revealed that JTD may regulate expressions of cytochrome c oxidase subunit 7C, metabotropic glutamate receptor 2, Slc30a1 zinc transporter 1, and apolipoprotein A-IV in VD mice and that their mechanisms involve biological processes like oxidative phosphorylation, regulation of neuron death, glutamate secretion, cellular ion homeostasis, and lipoprotein metabolism. Conclusions JTD may suppress VD development via multiple components, targets, and pathways. It may thus serve as a complementary treatment option for patients with VD.
Collapse
|
8
|
Liu XH, Zhou JT, Yan CX, Cheng C, Fan JN, Xu J, Zheng Q, Bai Q, Li Z, Li S, Li X. Single-cell RNA sequencing reveals a novel inhibitory effect of ApoA4 on NAFL mediated by liver-specific subsets of myeloid cells. Front Immunol 2022; 13:1038401. [PMID: 36426356 PMCID: PMC9678944 DOI: 10.3389/fimmu.2022.1038401] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/20/2022] [Indexed: 10/24/2023] Open
Abstract
The liver immune microenvironment is a key element in the development of hepatic inflammation in NAFLD. ApoA4 deficiency increases the hepatic lipid burden, insulin resistance, and metabolic inflammation. However, the effect of ApoA4 on liver immune cells and the precise immune cell subsets that exacerbate fatty liver remain elusive. The aim of this study was to profile the hepatic immune cells affected by ApoA4 in NAFL. We performed scRNA-seq on liver immune cells from WT and ApoA4-deficient mice administered a high-fat diet. Immunostaining and qRT-PCR analysis were used to validate the results of scRNA-seq. We identified 10 discrete immune cell populations comprising macrophages, DCs, granulocytes, B, T and NK&NKT cells and characterized their subsets, gene expression profiles, and functional modules. ApoA4 deficiency led to significant increases in the abundance of specific subsets, including inflammatory macrophages (2-Mφ-Cxcl9 and 4-Mφ-Cxcl2) and activated granulocytes (0-Gran-Wfdc17). Moreover, ApoA4 deficiency resulted in higher Lgals3, Ctss, Fcgr2b, Spp1, Cxcl2, and Elane levels and lower Nr4a1 levels in hepatic immune cells. These genes were consistent with human NAFLD-associated marker genes linked to disease severity. The expression of NE and IL-1β in granulocytes and macrophages as key ApoA4 targets were validate in the presence or absence of ApoA4 by immunostaining. The scRNA-seq data analyses revealed reprogramming of liver immune cells resulted from ApoA4 deficiency. We uncovered that the emergence of ApoA4-associated immune subsets (namely Cxcl9+ macrophage, Cxcl2+ macrophage and Wfdc17+ granulocyte), pathways, and NAFLD-related marker genes may promote the development of NAFL. These findings may provide novel therapeutic targets for NAFL and the foundations for further studying the effects of ApoA4 on immune cells in various diseases.
Collapse
Affiliation(s)
- Xiao-Huan Liu
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Jin-Ting Zhou
- Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi’an, China
| | - Chun-xia Yan
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Department of Pathology, Bio-Evidence Sciences Academy, The Western China Science and Technology Innovation Port, Xi’an Jiaotong University, Xi’an, China
| | - Cheng Cheng
- Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi’an, China
| | - Jing-Na Fan
- Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi’an, China
| | - Jing Xu
- Division of Endocrinology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Qiangsun Zheng
- Division of Cardiology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Qiang Bai
- Laboratory of Immunophysiology, GIGA Institute, Liège University, Liège, Belgium
| | - Zongfang Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Shengbin Li
- Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi’an, China
| | - Xiaoming Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
9
|
Cheng C, Liu XH, He J, Gao J, Zhou JT, Fan JN, Jin X, Zhang J, Chang L, Xiong Z, Yu J, Li S, Li X. Apolipoprotein A4 Restricts Diet-induced Hepatic Steatosis via SREBF1-mediated Lipogenesis and Enhances IRS-PI3K-Akt Signaling. Mol Nutr Food Res 2022; 66:e2101034. [PMID: 35909347 DOI: 10.1002/mnfr.202101034] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 07/03/2022] [Indexed: 11/12/2022]
Abstract
SCOPE Hepatic steatosis and insulin resistance (IR) are risk factors for many metabolic syndromes such as NAFLD and T2DM. ApoA4 improves glucose hemostasis by increasing glucose-stimulated insulin secretion and glucose uptake via PI3K-Akt activation in adipocytes. However, whether ApoA4 has an effect on hepatic steatosis or IR remains unclear. METHODS AND RESULTS ApoA4-knockout (KO) aggravates diet-induced obesity, hepatic steatosis and IR in mice promoted by increased hepatic lipogenesis gene expression based on RNA-seq data. Conversely, liver-specific overexpression of ApoA4 via AAV-ApoA4 transduction reverses the effect in ApoA4-KO mice, accompanied by suppressed hepatic lipogenesis, increased lipolysis, and fatty acid oxidation. Short-term treatment with recombinant ApoA4 protein improves glucose clearance and liver insulin sensitivity, and reduces hepatic lipogenesis gene expression in the absence of insulin. Moreover, in primary hepatocytes and a hepatic cell line, ApoA4 improves hepatic glucose uptake via IRS-PI3K-Akt signaling and decreases fat deposition and hepatic lipogenesis gene expression by inhibiting SREBF1 activity. CONCLUSION ApoA4 restricts hepatic steatosis by inhibiting SREBF1-mediated lipogenesis and improves insulin sensitivity and glucose uptake via IRS-PI3K-Akt signaling in the liver. These findings indicate that ApoA4 may serve as a therapeutic target for obesity-associated NAFLD. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Cheng Cheng
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Xiao-Huan Liu
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, Institute of Digestive Diseases. The Second Affiliated Hospital, Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Jing He
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Jing Gao
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Jin-Ting Zhou
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Jing-Na Fan
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Xi Jin
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Jianbo Zhang
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Liao Chang
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Zijun Xiong
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Jun Yu
- OneHealth Technology Company, Xi'an, 710000, China
| | - Shengbin Li
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Xiaoming Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, Institute of Digestive Diseases. The Second Affiliated Hospital, Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| |
Collapse
|
10
|
Iqbal Z, Fachim HA, Gibson JM, Baricevic-Jones I, Campbell AE, Geary B, Donn RP, Hamarashid D, Syed A, Whetton AD, Soran H, Heald AH. Changes in the Proteome Profile of People Achieving Remission of Type 2 Diabetes after Bariatric Surgery. J Clin Med 2021; 10:3659. [PMID: 34441954 PMCID: PMC8396849 DOI: 10.3390/jcm10163659] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
Bariatric surgery (BS) results in metabolic pathway recalibration. We have identified potential biomarkers in plasma of people achieving type 2 diabetes mellitus (T2DM) remission after BS. Longitudinal analysis was performed on plasma from 10 individuals following Roux-en-Y gastric bypass (n = 7) or sleeve gastrectomy (n = 3). Sequential window acquisition of all theoretical fragment ion spectra mass spectrometry (SWATH-MS) was done on samples taken at 4 months before (baseline) and 6 and 12 months after BS. Four hundred sixty-seven proteins were quantified by SWATH-MS. Principal component analysis resolved samples from distinct time points after selection of key discriminatory proteins: 25 proteins were differentially expressed between baseline and 6 months post-surgery; 39 proteins between baseline and 12 months. Eight proteins (SHBG, TF, PRG4, APOA4, LRG1, HSPA4, EPHX2 and PGLYRP) were significantly different to baseline at both 6 and 12 months post-surgery. The panel of proteins identified as consistently different included peptides related to insulin sensitivity (SHBG increase), systemic inflammation (TF and HSPA4-both decreased) and lipid metabolism (APOA4 decreased). We found significant changes in the proteome for eight proteins at 6- and 12-months post-BS, and several of these are key components in metabolic and inflammatory pathways. These may represent potential biomarkers of remission of T2DM.
Collapse
Affiliation(s)
- Zohaib Iqbal
- The School of Medicine and Manchester Academic Health Sciences Centre, Manchester University, Manchester M13 9PL, UK; (Z.I.); (J.M.G.); (R.P.D.); (H.S.)
- Department of Endocrinology, Diabetes and Metabolism, Salford Royal Foundation Trust, Salford M6 8HD, UK; (D.H.); (A.S.)
| | - Helene A. Fachim
- The School of Medicine and Manchester Academic Health Sciences Centre, Manchester University, Manchester M13 9PL, UK; (Z.I.); (J.M.G.); (R.P.D.); (H.S.)
- Department of Endocrinology, Diabetes and Metabolism, Salford Royal Foundation Trust, Salford M6 8HD, UK; (D.H.); (A.S.)
| | - J. Martin Gibson
- The School of Medicine and Manchester Academic Health Sciences Centre, Manchester University, Manchester M13 9PL, UK; (Z.I.); (J.M.G.); (R.P.D.); (H.S.)
- Department of Endocrinology, Diabetes and Metabolism, Salford Royal Foundation Trust, Salford M6 8HD, UK; (D.H.); (A.S.)
| | - Ivona Baricevic-Jones
- Stoller Biomarker Discovery Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (I.B.-J.); (A.E.C.); (B.G.); (A.D.W.)
| | - Amy E. Campbell
- Stoller Biomarker Discovery Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (I.B.-J.); (A.E.C.); (B.G.); (A.D.W.)
| | - Bethany Geary
- Stoller Biomarker Discovery Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (I.B.-J.); (A.E.C.); (B.G.); (A.D.W.)
| | - Rachelle P. Donn
- The School of Medicine and Manchester Academic Health Sciences Centre, Manchester University, Manchester M13 9PL, UK; (Z.I.); (J.M.G.); (R.P.D.); (H.S.)
| | - Dashne Hamarashid
- Department of Endocrinology, Diabetes and Metabolism, Salford Royal Foundation Trust, Salford M6 8HD, UK; (D.H.); (A.S.)
| | - Akheel Syed
- Department of Endocrinology, Diabetes and Metabolism, Salford Royal Foundation Trust, Salford M6 8HD, UK; (D.H.); (A.S.)
| | - Anthony D. Whetton
- Stoller Biomarker Discovery Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (I.B.-J.); (A.E.C.); (B.G.); (A.D.W.)
- Manchester National Institute for Health Research Biomedical Research Centre, Manchester M13 9WL, UK
| | - Handrean Soran
- The School of Medicine and Manchester Academic Health Sciences Centre, Manchester University, Manchester M13 9PL, UK; (Z.I.); (J.M.G.); (R.P.D.); (H.S.)
| | - Adrian H. Heald
- The School of Medicine and Manchester Academic Health Sciences Centre, Manchester University, Manchester M13 9PL, UK; (Z.I.); (J.M.G.); (R.P.D.); (H.S.)
- Department of Endocrinology, Diabetes and Metabolism, Salford Royal Foundation Trust, Salford M6 8HD, UK; (D.H.); (A.S.)
| |
Collapse
|
11
|
Zheng JJ, Agus JK, Hong BV, Tang X, Rhodes CH, Houts HE, Zhu C, Kang JW, Wong M, Xie Y, Lebrilla CB, Mallick E, Witwer KW, Zivkovic AM. Isolation of HDL by sequential flotation ultracentrifugation followed by size exclusion chromatography reveals size-based enrichment of HDL-associated proteins. Sci Rep 2021; 11:16086. [PMID: 34373542 PMCID: PMC8352908 DOI: 10.1038/s41598-021-95451-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 07/23/2021] [Indexed: 01/02/2023] Open
Abstract
High-density lipoprotein (HDL) particles have multiple beneficial and cardioprotective roles, yet our understanding of their full structural and functional repertoire is limited due to challenges in separating HDL particles from contaminating plasma proteins and other lipid-carrying particles that overlap HDL in size and/or density. Here we describe a method for isolating HDL particles using a combination of sequential flotation density ultracentrifugation and fast protein liquid chromatography with a size exclusion column. Purity was visualized by polyacrylamide gel electrophoresis and verified by proteomics, while size and structural integrity were confirmed by transmission electron microscopy. This HDL isolation method can be used to isolate a high yield of purified HDL from a low starting plasma volume for functional analyses. This method also enables investigators to select their specific HDL fraction of interest: from the least inclusive but highest purity HDL fraction eluting in the middle of the HDL peak, to pooling all of the fractions to capture the breadth of HDL particles in the original plasma sample. We show that certain proteins such as lecithin cholesterol acyltransferase (LCAT), phospholipid transfer protein (PLTP), and clusterin (CLUS) are enriched in large HDL particles whereas proteins such as alpha-2HS-glycoprotein (A2HSG), alpha-1 antitrypsin (A1AT), and vitamin D binding protein (VDBP) are enriched or found exclusively in small HDL particles.
Collapse
Affiliation(s)
| | - Joanne K Agus
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Brian V Hong
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Xinyu Tang
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | | | - Hannah E Houts
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Chenghao Zhu
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Jea Woo Kang
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Maurice Wong
- Department of Chemistry, University of California, Davis, Davis, CA, USA
| | - Yixuan Xie
- Department of Chemistry, University of California, Davis, Davis, CA, USA
| | - Carlito B Lebrilla
- Department of Chemistry, University of California, Davis, Davis, CA, USA
| | - Emily Mallick
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Angela M Zivkovic
- Department of Nutrition, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
12
|
Qu J, Fourman S, Fitzgerald M, Liu M, Nair S, Oses-Prieto J, Burlingame A, Morris JH, Davidson WS, Tso P, Bhargava A. Low-density lipoprotein receptor-related protein 1 (LRP1) is a novel receptor for apolipoprotein A4 (APOA4) in adipose tissue. Sci Rep 2021; 11:13289. [PMID: 34168225 PMCID: PMC8225859 DOI: 10.1038/s41598-021-92711-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 06/11/2021] [Indexed: 11/29/2022] Open
Abstract
Apolipoprotein A4 (APOA4) is one of the most abundant and versatile apolipoproteins facilitating lipid transport and metabolism. APOA4 is synthesized in the small intestine, packaged onto chylomicrons, secreted into intestinal lymph and transported via circulation to several tissues, including adipose. Since its discovery nearly 4 decades ago, to date, only platelet integrin αIIbβ3 has been identified as APOA4 receptor in the plasma. Using co-immunoprecipitation coupled with mass spectrometry, we probed the APOA4 interactome in mouse gonadal fat tissue, where ApoA4 gene is not transcribed but APOA4 protein is abundant. We demonstrate that lipoprotein receptor-related protein 1 (LRP1) is the cognate receptor for APOA4 in adipose tissue. LRP1 colocalized with APOA4 in adipocytes; it interacted with APOA4 under fasting condition and their interaction was enhanced during lipid feeding concomitant with increased APOA4 levels in plasma. In 3T3-L1 mature adipocytes, APOA4 promoted glucose uptake both in absence and presence of insulin in a dose-dependent manner. Knockdown of LRP1 abrogated APOA4-induced glucose uptake as well as activation of phosphatidylinositol 3 kinase (PI3K)-mediated protein kinase B (AKT). Taken together, we identified LRP1 as a novel receptor for APOA4 in promoting glucose uptake. Considering both APOA4 and LRP1 are multifunctional players in lipid and glucose metabolism, our finding opens up a door to better understand the molecular mechanisms along APOA4-LRP1 axis, whose dysregulation leads to obesity, cardiovascular disease, and diabetes.
Collapse
Affiliation(s)
- Jie Qu
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, 2180 E Galbraith Road, Cincinnati, 45237-0507, USA
| | - Sarah Fourman
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, 2180 E Galbraith Road, Cincinnati, 45237-0507, USA
| | - Maureen Fitzgerald
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, 2180 E Galbraith Road, Cincinnati, 45237-0507, USA
| | - Min Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, 2180 E Galbraith Road, Cincinnati, 45237-0507, USA
| | - Supna Nair
- Departments of Pharmaceutical Chemistry, University of California San Francisco, 600 16th Street, San Francisco, CA, 94158, USA
| | - Juan Oses-Prieto
- Departments of Pharmaceutical Chemistry, University of California San Francisco, 600 16th Street, San Francisco, CA, 94158, USA
| | - Alma Burlingame
- Departments of Pharmaceutical Chemistry, University of California San Francisco, 600 16th Street, San Francisco, CA, 94158, USA
| | - John H Morris
- Departments of Pharmaceutical Chemistry, University of California San Francisco, 600 16th Street, San Francisco, CA, 94158, USA
| | - W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, 2180 E Galbraith Road, Cincinnati, 45237-0507, USA
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, 2180 E Galbraith Road, Cincinnati, 45237-0507, USA
| | - Aditi Bhargava
- Department of Obstetrics and Gynecology, Center for Reproductive Sciences, University of California San Francisco, 513 Parnassus Avenue, Rm HSE1636, San Francisco, CA, 94143-0556, USA.
| |
Collapse
|
13
|
Tang D, Geng F, Yu C, Zhang R. Recent Application of Zebrafish Models in Atherosclerosis Research. Front Cell Dev Biol 2021; 9:643697. [PMID: 33718384 PMCID: PMC7947229 DOI: 10.3389/fcell.2021.643697] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/05/2021] [Indexed: 12/13/2022] Open
Abstract
Atherosclerotic cardiovascular disease is one of the leading causes of death worldwide. Establishing animal models of atherosclerosis is of great benefit for studying its complicated pathogenesis and screening and evaluating related drugs. Although researchers have generated a variety of models for atherosclerosis study in rabbits, mice and rats, the limitations of these models make it difficult to monitor the development of atherosclerosis, and these models are unsuitable for large scale screening of potential therapeutic targets. On the contrast, zebrafish can fulfill these purposes thanks to their fecundity, rapid development ex utero, embryonic transparency, and conserved lipid metabolism process. Thus, zebrafish have become a popular alternative animal model for atherosclerosis research. In this mini review, we summarize different zebrafish models used to study atherosclerosis, focusing on the latest applications of these models to the dynamic monitoring of atherosclerosis progression, mechanistic study of therapeutic intervention and drug screening, and assessment of the impacts of other risk factors.
Collapse
Affiliation(s)
- Dandan Tang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Fang Geng
- School of Life Sciences, Fudan University, Shanghai, China
| | - Chunxiao Yu
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Ruilin Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
14
|
Santana MFM, Lira ALA, Pinto RS, Minanni CA, Silva ARM, Sawada MIBAC, Nakandakare ER, Correa-Giannella MLC, Queiroz MS, Ronsein GE, Passarelli M. Enrichment of apolipoprotein A-IV and apolipoprotein D in the HDL proteome is associated with HDL functions in diabetic kidney disease without dialysis. Lipids Health Dis 2020; 19:205. [PMID: 32921312 PMCID: PMC7488728 DOI: 10.1186/s12944-020-01381-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/01/2020] [Indexed: 12/27/2022] Open
Abstract
Background and aims Diabetic kidney disease (DKD) is associated with lipid derangements that worsen kidney function and enhance cardiovascular (CVD) risk. The management of dyslipidemia, hypertension and other traditional risk factors does not completely prevent CVD complications, bringing up the participation of nontraditional risk factors such as advanced glycation end products (AGEs), carbamoylation and changes in the HDL proteome and functionality. The HDL composition, proteome, chemical modification and functionality were analyzed in nondialysis subjects with DKD categorized according to the estimated glomerular filtration rate (eGFR) and urinary albumin excretion rate (AER). Methods Individuals with DKD were divided into eGFR> 60 mL/min/1.73 m2 plus AER stages A1 and A2 (n = 10) and eGFR< 60 plus A3 (n = 25) and matched by age with control subjects (eGFR> 60; n = 8). Results Targeted proteomic analyses quantified 28 proteins associated with HDL in all groups, although only 2 were more highly expressed in the eGFR< 60 + A3 group than in the controls: apolipoprotein D (apoD) and apoA-IV. HDL from the eGFR< 60 + A3 group presented higher levels of total AGEs (20%), pentosidine (6.3%) and carbamoylation (4.2 x) and a reduced ability to remove 14C-cholesterol from macrophages (33%) in comparison to HDL from controls. The antioxidant role of HDL (lag time for LDL oxidation) was similar among groups, but HDL from the eGFR< 60 + A3 group presented a greater ability to inhibit the secretion of IL-6 and TNF-alpha (95%) in LPS-elicited macrophages in comparison to the control group. Conclusion The increase in apoD and apoA-IV could contribute to counteracting the HDL chemical modification by AGEs and carbamoylation, which contributes to HDL loss of function in well-established DKD.
Collapse
Affiliation(s)
- Monique F M Santana
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo 455, room 3305; CEP, São Paulo, 01246-000, Brazil
| | - Aécio L A Lira
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo 455, room 3305; CEP, São Paulo, 01246-000, Brazil
| | - Raphael S Pinto
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo 455, room 3305; CEP, São Paulo, 01246-000, Brazil.,Centro Universitário CESMAC, Maceio, Alagoas, Brazil
| | - Carlos A Minanni
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo 455, room 3305; CEP, São Paulo, 01246-000, Brazil.,Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein (HIAE), São Paulo, Brazil
| | - Amanda R M Silva
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Maria I B A C Sawada
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho, São Paulo, Brazil
| | - Edna R Nakandakare
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo 455, room 3305; CEP, São Paulo, 01246-000, Brazil
| | - Maria L C Correa-Giannella
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho, São Paulo, Brazil.,Laboratório de Carboidratos e Radioimunoensaio (LIM 18), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marcia S Queiroz
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho, São Paulo, Brazil
| | - Graziella E Ronsein
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Marisa Passarelli
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo 455, room 3305; CEP, São Paulo, 01246-000, Brazil. .,Programa de Pós-Graduação em Medicina, Universidade Nove de Julho, São Paulo, Brazil.
| |
Collapse
|
15
|
Apolipoprotein M: Research Progress and Clinical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:85-103. [PMID: 32705596 DOI: 10.1007/978-981-15-6082-8_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Apolipoprotein M (apoM) was first identified and characterized to the apolipoprotein family in 1999. Human apoM gene is located in a highly conserved segment in the major histocompatibility complex (MHC) class III locus on chromosome 6 and codes for an about 23 kDa protein that structurally belongs to the lipocalin superfamily. ApoM is selectively expressed in hepatocytes and in the tubular epithelium of kidney. In human plasma, apoM is mainly confined to the high-density lipoprotein (HDL) particles, but it may also occur in other lipoprotein classes, such as in the triglyceride-rich particles after fat intake. It has been demonstrated that apoM is critical for the formation of HDL, notably pre-beta HDL1. The antiatherogenic function of HDL is well established, and its ability to promote cholesterol efflux from foam cells in the atherosclerotic lesions is generally regarded as one of the key mechanisms behind this protective function. However, HDL could also display a variety of properties that may affect the complex atherosclerotic processes by other mechanisms, thus being involved in processes related to antioxidant defense, immune system, and systemic effects in septicemia, which may be partly contributed via its apolipoproteins and/or phospholipids. Moreover, it has been demonstrated that apoM functions as a natural carrier of sphingosin-1-phosphate (S1P) in vivo which may be related to its antiatherosclerotic and protective effects on endothelial cell barrier and anti-inflammatory properties. These may also provide a link between the diverse effects of HDL.
Collapse
|