1
|
Jirakran K, Vasupanrajit A, Tunvirachaisakul C, Almulla AF, Kubera M, Maes M. Lipid profiles in major depression, both with and without metabolic syndrome: associations with suicidal behaviors and neuroticism. BMC Psychiatry 2025; 25:379. [PMID: 40234788 PMCID: PMC11998271 DOI: 10.1186/s12888-025-06734-2] [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: 03/07/2024] [Accepted: 03/18/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Significant associations exist between major depressive disorder (MDD), metabolic syndrome (MetS), and cardiovascular disease, potentially attributable to heightened atherogenicity. This study aimed to ascertain if MDD, depression severity, suicidal behaviors, and neuroticism associate with elevated pro-atherogenic indices and reduced anti-atherogenic indices, including a reverse cholesterol transport (RCT) index. METHODS This study comprised 34 healthy controls and 33 MDD patients without MetS, and 35 controls and 31 MDD patients with MetS. It assessed total cholesterol (TC) and free cholesterol (FC), high-density lipoprotein cholesterol (HDLc), low-density lipoprotein cholesterol (LDLc), triglycerides (TG), apolipoprotein (ApoA), ApoB, cholesterol esterification rate, and a RCT composite. RESULTS No significant associations between MDD and lipids were seen in the total study group that combined individuals with and without MetS. In individuals devoid of MetS, MDD is significantly correlated with (a) elevated FC, TG, ApoB, Castelli risk index 1, and ApoB/ApoA, and (b) diminished HDLc, ApoA, and RCT index. In individuals without MetS, there are notable correlations between the severity of depression, suicidal tendencies, neuroticism, and ApoB/ApoA, Castelli risk, and RCT indices. CONCLUSIONS The link between lipids and MDD features cannot be adequately estimated by combining participants with and without MetS. It should be examined in a study sample that excludes subjects with MetS. The depression phenome, suicidal behaviors, and neuroticism correlate with diminished RCT and heightened atherogenicity, which are likely implicated in the pathophysiology of MDD. Increased atherogenicity and lowered RCT may represent novel drug targets for the treatment and prevention of MDD, neuroticism, and suicidal behaviors.
Collapse
Affiliation(s)
- Ketsupar Jirakran
- School of Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, 610072, China
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the, Thai Red Cross Societyaq , Bangkok, Thailand
- Department of Pediatric, Faculty of Medicine, Center of Excellence for Maximizing Children's Developmental Potential, Chulalongkorn University, Bangkok, Thailand
| | - Asara Vasupanrajit
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the, Thai Red Cross Societyaq , Bangkok, Thailand
| | - Chavit Tunvirachaisakul
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the, Thai Red Cross Societyaq , Bangkok, Thailand.
- Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| | - Abbas F Almulla
- School of Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, 610072, China
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the, Thai Red Cross Societyaq , Bangkok, Thailand
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Marta Kubera
- Department of Experimental Neuroendocrinology, Laboratory of Immunoendocrinology, Polish Academy of Sciences, Maj Institute of Pharmacology, Smetna 12, Krakow, 31-343, Poland
| | - Michael Maes
- School of Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, 610072, China.
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the, Thai Red Cross Societyaq , Bangkok, Thailand.
- Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
- Research Center, Medical University of Plovdiv, Plovdiv, Bulgaria.
- Research and Innovation Program for the Development of MU - PLOVDIV- (SRIPD-MUP), Creation of a network of research higher schools, National plan for recovery and sustainability, European Union - NextGeneration, EU, Maastricht, Netherlands.
- Kyung Hee University, 26 Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Korea.
| |
Collapse
|
2
|
Li Y, Feng Q, Wang L, Gao X, Xi Y, Ye L, Ji J, Yang X, Zhai G. Current targeting strategies and advanced nanoplatforms for atherosclerosis therapy. J Drug Target 2024; 32:128-147. [PMID: 38217526 DOI: 10.1080/1061186x.2023.2300694] [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: 09/16/2023] [Accepted: 12/24/2023] [Indexed: 01/15/2024]
Abstract
Atherosclerosis is one of the major causes of death worldwide, and it is closely related to many cardiovascular diseases, such as stroke, myocardial infraction and angina. Although traditional surgical and pharmacological interventions can effectively retard or slow down the progression of atherosclerosis, it is very difficult to prevent or even reverse this disease. In recent years, with the rapid development of nanotechnology, various nanoagents have been designed and applied to different diseases including atherosclerosis. The unique atherosclerotic microenvironment with signature biological components allows nanoplatforms to distinguish atherosclerotic lesions from normal tissue and to approach plaques specifically. Based on the process of atherosclerotic plaque formation, this review summarises the nanodrug delivery strategies for atherosclerotic therapy, trying to provide help for researchers to understand the existing atherosclerosis management approaches as well as challenges and to reasonably design anti-atherosclerotic nanoplatforms.
Collapse
Affiliation(s)
- Yingchao Li
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Qixiang Feng
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Luyue Wang
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Xi Gao
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Yanwei Xi
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Lei Ye
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Jianbo Ji
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Xiaoye Yang
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Guangxi Zhai
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
3
|
Andraski AB, Sacks FM, Aikawa M, Singh SA. Understanding HDL Metabolism and Biology Through In Vivo Tracer Kinetics. Arterioscler Thromb Vasc Biol 2024; 44:76-88. [PMID: 38031838 PMCID: PMC10842918 DOI: 10.1161/atvbaha.123.319742] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023]
Abstract
HDL (high-density lipoprotein), owing to its high protein content and small size, is the densest circulating lipoprotein. In contrast to lipid-laden VLDL (very-low-density lipoprotein) and LDL (low-density lipoprotein) that promote atherosclerosis, HDL is hypothesized to mitigate atherosclerosis via reverse cholesterol transport, a process that entails the uptake and clearance of excess cholesterol from peripheral tissues. This process is mediated by APOA1 (apolipoprotein A-I), the primary structural protein of HDL, as well as by the activities of additional HDL proteins. Tracer-dependent kinetic studies are an invaluable tool to study HDL-mediated reverse cholesterol transport and overall HDL metabolism in humans when a cardiovascular disease therapy is investigated. Unfortunately, HDL cholesterol-raising therapies have not been successful at reducing cardiovascular events suggesting an incomplete picture of HDL biology. However, as HDL tracer studies have evolved from radioactive isotope- to stable isotope-based strategies that in turn are reliant on mass spectrometry technologies, the complexity of the HDL proteome and its metabolism can be more readily addressed. In this review, we outline the motivations, timelines, advantages, and disadvantages of the various tracer kinetics strategies. We also feature the metabolic properties of select HDL proteins known to regulate reverse cholesterol transport, which in turn underscore that HDL lipoproteins comprise a heterogeneous particle population whose distinct protein constituents and kinetics likely determine its function and potential contribution to cholesterol clearance.
Collapse
Affiliation(s)
- Allison B. Andraski
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Frank M. Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Sasha A. Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
4
|
Tao Y, Lan X, Zhang Y, Fu C, Liu L, Cao F, Guo W. Biomimetic nanomedicines for precise atherosclerosis theranostics. Acta Pharm Sin B 2023; 13:4442-4460. [PMID: 37969739 PMCID: PMC10638499 DOI: 10.1016/j.apsb.2022.11.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/13/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis (AS) is a leading cause of the life-threatening cardiovascular disease (CVD), creating an urgent need for efficient, biocompatible therapeutics for diagnosis and treatment. Biomimetic nanomedicines (bNMs) are moving closer to fulfilling this need, pushing back the frontier of nano-based drug delivery systems design. This review seeks to outline how these nanomedicines (NMs) might work to diagnose and treat atherosclerosis, to trace the trajectory of their development to date and in the coming years, and to provide a foundation for further discussion about atherosclerotic theranostics.
Collapse
Affiliation(s)
- Ying Tao
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Biomedical Engineering & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Xinmiao Lan
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Yang Zhang
- Department of Cardiology, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Chenxing Fu
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Lu Liu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR 999077, China
| | - Feng Cao
- Department of Cardiology, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Weisheng Guo
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Biomedical Engineering & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| |
Collapse
|
5
|
Almulla AF, Thipakorn Y, Algon AAA, Tunvirachaisakul C, Al-Hakeim HK, Maes M. Reverse cholesterol transport and lipid peroxidation biomarkers in major depression and bipolar disorder: A systematic review and meta-analysis. Brain Behav Immun 2023; 113:374-388. [PMID: 37557967 DOI: 10.1016/j.bbi.2023.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/01/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Major depression (MDD) and bipolar disorder (BD) are linked to immune activation, increased oxidative stress, and lower antioxidant defenses. OBJECTIVES To systematically review and meta-analyze all data concerning biomarkers of reverse cholesterol transport (RCT), lipid-associated antioxidants, lipid peroxidation products, and autoimmune responses to oxidatively modified lipid epitopes in MDD and BD. METHODS Databases including PubMed, Google scholar and SciFinder were searched to identify eligible studies from inception to January 10th, 2023. Guidelines of Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed. RESULTS The current meta-analysis included 176 studies (60 BD and 116 MDD) and examined 34,051 participants, namely 17,094 with affective disorders and 16,957 healthy controls. Patients with MDD and BD showed a) significantly decreased RCT (mainly lowered high-density lipoprotein cholesterol and paraoxonase 1); b) lowered lipid soluble vitamins (including vitamin A, D, and coenzyme Q10); c) increased lipid peroxidation and aldehyde formation, mainly increased malondialdehyde (MDA), 4-hydroxynonenal, peroxides, and 8-isoprostanes; and d) Immunoglobulin (Ig)G responses to oxidized low-density lipoprotein and IgM responses to MDA. The ratio of all lipid peroxidation biomarkers/all lipid-associated antioxidant defenses was significantly increased in MDD (standardized mean difference or SMD = 0.433; 95% confidence intervals (CI): 0.312; 0.554) and BD (SMD = 0.653; CI: 0.501-0.806). This ratio was significantly greater in BD than MDD (p = 0.027). CONCLUSION In MDD/BD, lowered RCT, a key antioxidant and anti-inflammatory pathway, may drive increased lipid peroxidation, aldehyde formation, and autoimmune responses to oxidative specific epitopes, which all together cause increased immune-inflammatory responses and neuro-affective toxicity.
Collapse
Affiliation(s)
- Abbas F Almulla
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Yanin Thipakorn
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| | | | - Chavit Tunvirachaisakul
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria; Research Institute, Medical University in Plovdiv, Plovdiv, Bulgaria; Department of Psychiatry, IMPACT Strategic Research Centre, Deakin University, Geelong, Victoria, Australia; Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China.
| |
Collapse
|
6
|
Rein-Fischboeck L, Pohl R, Haberl EM, Mages W, Girke P, Liebisch G, Krautbauer S, Buechler C. Lower adiposity does not protect beta-2 syntrophin null mice from hepatic steatosis and inflammation in experimental non-alcoholic steatohepatitis. Gene 2023; 859:147209. [PMID: 36681100 DOI: 10.1016/j.gene.2023.147209] [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: 06/17/2022] [Revised: 12/21/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
Visceral adiposity is strongly associated with liver steatosis, which predisposes to the development of non-alcoholic steatohepatitis (NASH). Mice with loss of the molecular adapter protein beta-2 syntrophin (SNTB2) have greatly reduced intra-abdominal fat mass. Hepatic expression of proteins with a role in fatty acid metabolism such as fatty acid synthase was nevertheless normal. This was also the case for proteins regulating cholesterol synthesis and uptake. Yet, a slight induction of hepatic cholesterol was noticed in the mutant mice. When mice were fed a methionine choline deficient (MCD) diet to induce NASH, liver cholesteryl ester content was induced in the wild type but not the mutant mice. Serum cholesterol of the mice fed a MCD diet declined and this was significant for the SNTB2 null mice. Though the mutant mice lost less fat mass than the wild type animals, hepatic triglyceride levels were similar between the groups. Proteins involved in fatty acid or cholesterol metabolism such as fatty acid synthase, apolipoprotein E and low-density lipoprotein receptor did not differ between the genotypes. Hepatic oxidative stress and liver inflammation of mutant and wild type mice were comparable. Mutant mice had lower hepatic levels of secondary bile acids and higher cholesterol storage in epididymal fat, and this may partly prevent hepatic cholesterol deposition. In summary, the current study shows that SNTB2 null mice have low intra-abdominal fat mass and do not accumulate hepatic cholesteryl esters when fed a MCD diet. Nevertheless, the SNTB2 null mice develop a similar NASH pathology as wild type mice suggesting a minor role of intra-abdominal fat and liver cholesteryl esters in this model.
Collapse
Affiliation(s)
- Lisa Rein-Fischboeck
- Department of Internal Medicine I, Regensburg University Hospital, D-93053 Regensburg, Germany
| | - Rebekka Pohl
- Department of Internal Medicine I, Regensburg University Hospital, D-93053 Regensburg, Germany
| | - Elisabeth M Haberl
- Department of Internal Medicine I, Regensburg University Hospital, D-93053 Regensburg, Germany
| | - Wolfgang Mages
- Department of Genetics, University of Regensburg, D-93040 Regensburg, Germany
| | - Philipp Girke
- Department of Genetics, University of Regensburg, D-93040 Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, D-93053 Regensburg, Germany
| | - Sabrina Krautbauer
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, D-93053 Regensburg, Germany
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, D-93053 Regensburg, Germany.
| |
Collapse
|
7
|
OxLDL-Induced Foam Cell Formation Inhibitory Activity of Pepsin Hydrolysate of Ark Shell (Scapharca subcrenata (Lischke, 1869)) in RAW264.7 Macrophages. J Food Biochem 2023. [DOI: 10.1155/2023/6905673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Inhibitory effect of ark shell (Scapharca subcrenata (Lischke, 1869)) proteolytic hydrolysates (ASHs) on oxidized low-density lipoprotein (oxLDL)-induced macrophage foam cell formation was investigated. Two types of ASHs were prepared by Alcalase® and pepsin, ASAH (ark shell-Alcalase® hydrolysates), and ASPH (ark shell-pepsin hydrolysate). Oil Red O staining results showed that ASPH suppressed foam cell formation and lipid accumulation more than ASAH in oxLDL-induced foam cell formation of RAW264.7 macrophages. ASPH reduced the levels of total cholesterol, cholesterol ester, and free cholesterol in oxLDL-treated RAW264.7 macrophages. It was found that ASPH increased cholesterol efflux and decreased cholesterol influx rate. In this regard, protein expressions of CD36 and scavenger receptor class A1 (SR-A1) for cholesterol influx and ATP-binding cassette transporter A1 and G1 (ABCA1 and ABCG1) for cholesterol efflux were investigated. ASPH treatment resulted in an increase of ABCA1 and ABCG1 expression but downregulated CD36 and SR-A1 expression. Furthermore, ASPH suppressed production of proinflammatory cytokines, including tumor necrosis factor-α and interleukin-6 and -1β, through regulating nuclear factor-kappa B (NF-κB) in oxLDL-induced foam cell formation of RAW264.7 macrophages. Taken together, our data indicate that ASPH might be a useful ingredient in functional foods for ameliorating atherosclerosis by preventing foam cell formation.
Collapse
|
8
|
Yang J, Chen Y, Zou T, Xue B, Yang F, Wang X, Huo Y, Yan B, Xu Y, He S, Yin Y, Wang J, Zhu X, Zhang L, Zhou Y, Tai Z, Shuai P, Yu M, Luo Q, Cheng Y, Gong B, Zhu X, Zhang J, Sun X, Lin Y, Zhang H, Yang Z. Cholesterol homeostasis regulated by ABCA1 is critical for retinal ganglion cell survival. SCIENCE CHINA. LIFE SCIENCES 2023; 66:211-225. [PMID: 35829808 DOI: 10.1007/s11427-021-2126-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 06/17/2022] [Indexed: 10/17/2022]
Abstract
Genome-wide association studies have suggested a link between primary open-angle glaucoma and the function of ABCA1. ABCA1 is a key regulator of cholesterol efflux and the biogenesis of high-density lipoprotein (HDL) particles. Here, we showed that the POAG risk allele near ABCA1 attenuated ABCA1 expression in cultured cells. Consistently, POAG patients exhibited lower ABCA1 expression, reduced HDL, and higher cholesterol in white blood cells. Ablation of Abca1 in mice failed to form HDL, leading to elevated cholesterol levels in the retina. Counting retinal ganglion cells (RGCs) by using an artificial intelligence (AI) program revealed that Abca1-deficient mice progressively lost RGCs with age. Single-cell RNA sequencing (scRNA-seq) revealed aberrant oxidative phosphorylation in the Abca1-/- retina, as well as activation of the mTORC1 signaling pathway and suppression of autophagy. Treatment of Abca1-/- mice using atorvastatin reduced the cholesterol level in the retina, thereby improving metabolism and protecting RGCs from death. Collectively, we show that lower ABCA1 expression and lower HDL are risk factors for POAG. Accumulated cholesterol in the Abca1-/- retina causes profound aberrant metabolism, leading to a POAG-like phenotype that can be prevented by atorvastatin. Our findings establish statin use as a preventive treatment for POAG associated with lower ABCA1 expression.
Collapse
Affiliation(s)
- Jialiang Yang
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yuhong Chen
- Department of Ophthalmology and Visual Science, Eye and Ear Nose Throat (ENT) Hospital, Fudan University, Shanghai, 200032, China
| | - Tongdan Zou
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Bai Xue
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Fang Yang
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Xiangzhou Wang
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Yibo Huo
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Boyun Yan
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Yuxia Xu
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Shiyu He
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yi Yin
- Institute of Chengdu Biology, Sichuan Translational Medicine Hospital, Chinese Academy of Sciences, Chengdu, 610093, China
| | - Jing Wang
- Institute of Chengdu Biology, Sichuan Translational Medicine Hospital, Chinese Academy of Sciences, Chengdu, 610093, China
| | - Xiong Zhu
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lin Zhang
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yu Zhou
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Zhengfu Tai
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Ping Shuai
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Man Yu
- Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Qian Luo
- Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yilian Cheng
- Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Bo Gong
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Xianjun Zhu
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Jing Zhang
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xinghuai Sun
- Department of Ophthalmology and Visual Science, Eye and Ear Nose Throat (ENT) Hospital, Fudan University, Shanghai, 200032, China
| | - Ying Lin
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Houbin Zhang
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Zhenglin Yang
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Institute of Chengdu Biology, Sichuan Translational Medicine Hospital, Chinese Academy of Sciences, Chengdu, 610093, China.
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072, China.
| |
Collapse
|
9
|
Cyclodextrin boostered-high density lipoprotein for antiatherosclerosis by regulating cholesterol efflux and efferocytosis. Carbohydr Polym 2022; 292:119632. [DOI: 10.1016/j.carbpol.2022.119632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/07/2022] [Accepted: 05/14/2022] [Indexed: 02/05/2023]
|
10
|
Noveir SD, Kerman BE, Xian H, Meuret C, Smadi S, Martinez AE, Johansson J, Zetterberg H, Parks BA, Kuklenyik Z, Mack WJ, Johansson JO, Yassine HN. Effect of the ABCA1 agonist CS-6253 on amyloid-β and lipoprotein metabolism in cynomolgus monkeys. Alzheimers Res Ther 2022; 14:87. [PMID: 35751102 PMCID: PMC9229758 DOI: 10.1186/s13195-022-01028-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 06/07/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Inducing brain ATP-binding cassette 1 (ABCA1) activity in Alzheimer's disease (AD) mouse models is associated with improvement in AD pathology. The purpose of this study was to investigate the effects of the ABCA1 agonist peptide CS-6253 on amyloid-β peptides (Aβ) and lipoproteins in plasma and cerebrospinal fluid (CSF) of cynomolgus monkeys, a species with amyloid and lipoprotein metabolism similar to humans. METHODS CS-6253 peptide was injected intravenously into cynomolgus monkeys at various doses in three different studies. Plasma and CSF samples were collected at several time points before and after treatment. Levels of cholesterol, triglyceride (TG), lipoprotein particles, apolipoproteins, and Aβ were measured using ELISA, ion-mobility analysis, and asymmetric-flow field-flow fractionation (AF4). The relationship between the change in levels of these biomarkers was analyzed using multiple linear regression models and linear mixed-effects models. RESULTS Following CS-6253 intravenous injection, within minutes, small plasma high-density lipoprotein (HDL) particles were increased. In two independent experiments, plasma TG, apolipoprotein E (apoE), and Aβ42/40 ratio were transiently increased following CS-6253 intravenous injection. This change was associated with a non-significant decrease in CSF Aβ42. Both plasma total cholesterol and HDL-cholesterol levels were reduced following treatment. AF4 fractionation revealed that CS-6253 treatment displaced apoE from HDL to intermediate-density- and low density-lipoprotein (IDL/LDL)-sized particles in plasma. In contrast to plasma, CS-6253 had no effect on the assessed CSF apolipoproteins or lipids. CONCLUSIONS Treatment with the ABCA1 agonist CS-6253 appears to favor Aβ clearance from the brain.
Collapse
Affiliation(s)
- Sasan D Noveir
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Bilal E Kerman
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Haotian Xian
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Cristiana Meuret
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Sabrina Smadi
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Ashley E Martinez
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Bryan A Parks
- Centers for Disease Control and Prevention, Atlanta, GA, 30341, USA
| | | | - Wendy J Mack
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, 90033, USA
| | | | - Hussein N Yassine
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
11
|
Ghaffari T, Moradi N, Chamani E, Ebadi Z, Fadaei R, Alizadeh-Fanalou S, Yarahmadi S, Fallah S. Captopril and Spironolactone Can Attenuate Diabetic Nephropathy in Wistar Rats by Targeting ABCA1 and microRNA-33. Curr Pharm Des 2022; 28:1367-1372. [DOI: 10.2174/1381612828666220401143249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/24/2022] [Indexed: 11/22/2022]
Abstract
Background:
Nephropathy diabetes is one of the important causes of death and a more prevalent cause of end-stage renal disease.
Objective:
The present study investigated the effect of applying spironolactone and captopril and their combination on some renal performance indices and cholesterol-efflux-related gene expression in nephropathy diabetic rats.
Methods:
Intraperitoneal injection of streptozotocin was used to induce diabetes in rats. FBS, creatinine, and BUN were assayed using the calorimetry technique; also, urine microalbumin was assayed by ELISA. Hepatic gene expressions of ABCA1, ABCG1, and miR-33 were evaluated by the real-time PCR method.
Results:
FBS levels in the captopril-treated group were significantly decreased compared with the untreated diabetic group. BUN levels of treated groups with captopril and a combination of captopril + spironolactone were significantly increased. GFR of both treated diabetic groups with captopril and spironolactone was significantly lower than an untreated diabetic group. ABCA1 gene expression in hepatic cells of the combination of spironolactone + captopril treated group was significantly increased compared to other treated and untreated diabetic groups. The hepatic expression of the ABCG1 gene in the treated and untreated diabetic groups was significantly lower than in the control group. Treatment of the diabetic group with only combination therapy decreased the hepatic gene expression of miR-33 significantly.
Conclusion:
Obtained results suggest that S+C combination therapy can improve nephropathy and diabetes disorders by targeting the ABCA1 and miR-33 gene expression. It is suggested miR-33 and ABCA1 genes evaluation could be a new therapeutic strategy for nephropathy diabetes remediation.
Collapse
Affiliation(s)
- Tina Ghaffari
- Department of Biochemistry and Nutrition, School of Medicine Iran University of Medical Sciences
| | - Nariman Moradi
- Department of Clinical Biochemistry, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Elham Chamani
- Department of Clinical Biochemistry, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Zahra Ebadi
- Department of Biochemistry and Nutrition, School of Medicine Iran University of Medical Sciences
| | - Reza Fadaei
- Sleep Disorders Research Center, Research Institute for Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shahin Alizadeh-Fanalou
- Department of Biochemistry and Nutrition, School of Medicine Iran University of Medical Sciences
| | - Sahar Yarahmadi
- Department of Biochemistry and Nutrition, School of Medicine Iran University of Medical Sciences
| | - Soudabeh Fallah
- Department of Biochemistry and Nutrition, School of Medicine Iran University of Medical Sciences
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW Dietary fat compared to carbohydrate increases the plasma concentration of high-density lipoprotein (HDL)-cholesterol. However, neither the mechanism nor its connection to cardiovascular disease is known. RECENT FINDINGS Protein-based subspecies of HDL, especially those containing apolipoprotein E (apoE) or apolipoprotein C3 (apoC3), offer a glimpse of a vast metabolic system related to atherogenicity, coronary heart disease (CHD) and other diseases. ApoE stimulates several processes that define reverse cholesterol transport through HDL, specifically secretion of active HDL subspecies, cholesterol efflux to HDL from macrophages involved in atherogenesis, size enlargement of HDL with cholesterol ester, and rapid clearance from the circulation. Dietary unsaturated fat stimulates the flux of HDL that contains apoE through these protective pathways. Effective reverse cholesterol transport may lessen atherogenesis and prevent disease. In contrast, apoC3 abrogates the benefit of apoE on reverse cholesterol transport, which may account for the association of HDL that contains apoC3 with dyslipidemia, obesity and CHD. SUMMARY Dietary unsaturated fat and carbohydrate affect the metabolism of protein-defined HDL subspecies containing apoE or apoC3 accelerating or retarding reverse cholesterol transport, thus demonstrating new mechanisms that may link diet to HDL and to CHD.
Collapse
Affiliation(s)
- Frank M. Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
13
|
Sunidhi S, Sacher S, Atul, Garg P, Ray A. Elucidating the Structural Features of ABCA1 in its Heterogeneous Membrane Environment. Front Mol Biosci 2022; 8:803078. [PMID: 35155567 PMCID: PMC8830745 DOI: 10.3389/fmolb.2021.803078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/30/2021] [Indexed: 11/17/2022] Open
Abstract
ATP Binding Cassette Transporter A1 (ABCA1) plays an integral part in Reverse Cholesterol Transport (RCT) and is critical for maintaining lipid homeostasis. One theory of lipid efflux by the transporter (alternating access) proposes that ABCA1 harbours two different conformations that provide alternating access for lipid binding and release. This is followed by sequestration via a direct interaction between ABCA1 and its partner, ApoA1. The other theory (lateral access) proposes that ABCA1 obtains lipids laterally from the membrane to form a temporary extracellular “reservoir”. This reservoir contains an isolated lipid monolayer due to the net accumulation of lipids in the exofacial leaflet. Recently, a full-length Cryo-EM structure of this 2,261-residue transmembrane protein showed its discreetly folded domains and have detected the presence of a tunnel enclosed within the extracellular domains (ECDs) but not in the TMDs, giving it an outward-facing conformation. This structure was hypothesized to substantiate the lateral access theory. Utilizing long time-scale multiple replica atomistic molecular dynamics simulations (MDS), we simulated the structure in a large heterogeneous lipid environment and found that the protein undergoes several large conformational changes in its extremities. We observed that the cavity enclosed within ATP unbound form of ABCA1 is narrow at the distal ends of TMD as well as the ECD region substantiating the “lateral access” theory. We have also characterized ABCA1 and the lipid dynamics along with the protein-lipid interactions in the heterogeneous environment, providing novel insights into understanding ABCA1 conformation at an atomistic level.
Collapse
|
14
|
Morris G, Puri BK, Bortolasci CC, Carvalho A, Berk M, Walder K, Moreira EG, Maes M. The role of high-density lipoprotein cholesterol, apolipoprotein A and paraoxonase-1 in the pathophysiology of neuroprogressive disorders. Neurosci Biobehav Rev 2021; 125:244-263. [PMID: 33657433 DOI: 10.1016/j.neubiorev.2021.02.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 01/29/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022]
Abstract
Lowered high-density lipoprotein (HDL) cholesterol has been reported in major depressive disorder, bipolar disorder, first episode of psychosis, and schizophrenia. HDL, its major apolipoprotein component, ApoA1, and the antioxidant enzyme paraoxonase (PON)1 (which is normally bound to ApoA1) all have anti-atherogenic, antioxidant, anti-inflammatory, and immunomodulatory roles, which are discussed in this paper. The paper details the pathways mediating the anti-inflammatory effects of HDL, ApoA1 and PON1 and describes the mechanisms leading to compromised HDL and PON1 levels and function in an environment of chronic inflammation. The molecular mechanisms by which changes in HDL, ApoA1 and PON1 might contribute to the pathophysiology of the neuroprogressive disorders are explained. Moreover, the anti-inflammatory actions of ApoM-mediated sphingosine 1-phosphate (S1P) signalling are reviewed as well as the deleterious effects of chronic inflammation and oxidative stress on ApoM/S1P signalling. Finally, therapeutic interventions specifically aimed at improving the levels and function of HDL and PON1 while reducing levels of inflammation and oxidative stress are considered. These include the so-called Mediterranean diet, extra virgin olive oil, polyphenols, flavonoids, isoflavones, pomegranate juice, melatonin and the Mediterranean diet combined with the ketogenic diet.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | | | - Chiara C Bortolasci
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia.
| | - Andre Carvalho
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Michael Berk
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Ken Walder
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia
| | - Estefania G Moreira
- Post-Graduation Program in Health Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Michael Maes
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
15
|
Cardiac apoptosis caused by elevated cholesterol level in experimental autoimmune myocarditis. Exp Cell Res 2020; 395:112169. [PMID: 32653410 DOI: 10.1016/j.yexcr.2020.112169] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 11/21/2022]
Abstract
It has been reported that cholesterol-lowing agents can ameliorate severity of myocarditis. However, the beneficial effect of the agents has been claimed to be independent of cholesterol reduction as there is no significant change in the plasma cholesterol level in myocarditis. In the present study, using experimental autoimmune myocarditis (EAM) rats as an animal model, we demonstrated that EAM induced elevation of cholesterol level and impaired cholesterol efflux capacity in the cardiac tissue. Moreover, serum high-density lipoprotein (HDL) content was reduced and HDL function associated protein Paraoxonase 1 (PON1) activity was decreased. Besides, the major structural protein within HDL, Apolipoprotein A1 (ApoA1) expression in the cardiac tissues was significantly reduced while the level of serum ApoA1 was not significantly altered. Importantly, cholesterol depleting agent methyl-β-cyclodextrin (MβCD) alleviated the development of EAM, as monitored by decreased ratio of heart weight to body weight (HW/BW), decreased infiltration of inflammatory cells and collagen deposition, improved cardiac function, reduced expression of apoptosis-related protein Bax, Fas, FasL and caspase-3 and increased level of anti-apoptotic protein Bcl-2. These results suggest that reduction of cholesterol level in cardiac tissue could suppress EAM-induced cardiac apoptosis through both intrinsic and extrinsic apoptotic pathways.
Collapse
|
16
|
Cholesterol uptake and efflux are impaired in human trophoblast cells from pregnancies with maternal supraphysiological hypercholesterolemia. Sci Rep 2020; 10:5264. [PMID: 32210256 PMCID: PMC7093446 DOI: 10.1038/s41598-020-61629-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/17/2020] [Indexed: 12/14/2022] Open
Abstract
Maternal physiological (MPH) or supraphysiological hypercholesterolaemia (MSPH) occurs during pregnancy. Cholesterol trafficking from maternal to foetal circulation requires the uptake of maternal LDL and HDL by syncytiotrophoblast and cholesterol efflux from this multinucleated tissue to ApoA-I and HDL. We aimed to determine the effects of MSPH on placental cholesterol trafficking. Placental tissue and primary human trophoblast (PHT) were isolated from pregnant women with total cholesterol <280 md/dL (MPH, n = 27) or ≥280 md/dL (MSPH, n = 28). The lipid profile in umbilical cord blood from MPH and MSPH neonates was similar. The abundance of LDL receptor (LDLR) and HDL receptor (SR-BI) was comparable between MSPH and MPH placentas. However, LDLR was localized mainly in the syncytiotrophoblast surface and was associated with reduced placental levels of its ligand ApoB. In PHT from MSPH, the uptake of LDL and HDL was lower compared to MPH, without changes in LDLR and reduced levels of SR-BI. Regarding cholesterol efflux, in MSPH placentas, the abundance of cholesterol transporter ABCA1 was increased, while ABCG1 and SR-BI were reduced. In PHT from MSPH, the cholesterol efflux to ApoA-I was increased and to HDL was reduced, along with reduced levels of ABCG1, compared to MPH. Inhibition of SR-BI did not change cholesterol efflux in PHT. The TC content in PHT was comparable in MPH and MSPH cells. However, free cholesterol was increased in MSPH cells. We conclude that MSPH alters the trafficking and content of cholesterol in placental trophoblasts, which could be associated with changes in the placenta-mediated maternal-to-foetal cholesterol trafficking.
Collapse
|
17
|
D-Allulose enhances uptake of HDL-cholesterol into rat's primary hepatocyte via SR-B1. Cytotechnology 2020; 72:295-301. [PMID: 32086695 DOI: 10.1007/s10616-020-00378-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/24/2020] [Accepted: 02/13/2020] [Indexed: 10/25/2022] Open
Abstract
D-Allulose, a C-3 epimer of D-fructose, is a rare sugar and a non-caloric sweetener. D-Allulose is reported to have several health benefits, such as suppressing a rise in postprandial glucose levels and preventing fat accumulation in rodents and humans. Additionally, low HDL-cholesterol levels post-D-allulose feeding were observed in humans but it is unclear how D-allulose decreased HDL-cholesterol levels. It is necessary to research the mechanism of HDL-cholesterol reduction by D-allulose ingestion because low HDL-cholesterol levels are known to associate with increased atherosclerosis risk. We therefore investigated the mechanism by which D-allulose lowers HDL-cholesterol using rat's primary hepatocytes. Sprague Dawley rats were fed an AIN-93G based diet containing 3% D-allulose for 2 weeks. Thereafter, primary hepatocytes were isolated by perfusion of collagenase. We measured the ability of HDL-cholesterol uptake in hepatocytes and the protein levels of scavenger receptor class B type 1 (SR-B1) as a HDL-cholesterol receptor. D-Allulose enhanced hepatocyte uptake of HDL-cholesterol, with a concurrent increase in hepatic SR-B1 protein levels. The results suggest that D-allulose enhances HDL-cholesterol uptake into the liver by increasing SR-B1 expression. It is estimated that HDL-cholesterol levels decreased accordingly. Since SR-B1 overexpression would decrease HDL-cholesterol levels, reportedly preventing atherosclerosis development, D-allulose could be a useful sweetener for atherosclerosis prevention.
Collapse
|
18
|
Ji A, Wang X, Noffsinger VP, Jennings D, de Beer MC, de Beer FC, Tannock LR, Webb NR. Serum amyloid A is not incorporated into HDL during HDL biogenesis. J Lipid Res 2020; 61:328-337. [PMID: 31915139 DOI: 10.1194/jlr.ra119000329] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 01/06/2020] [Indexed: 11/20/2022] Open
Abstract
Liver-derived serum amyloid A (SAA) is present in plasma where it is mainly associated with HDL and from which it is cleared more rapidly than are the other major HDL-associated apolipoproteins. Although evidence suggests that lipid-free and HDL-associated forms of SAA have different activities, the pathways by which SAA associates and disassociates with HDL are poorly understood. In this study, we investigated SAA lipidation by hepatocytes and how this lipidation relates to the formation of nascent HDL particles. We also examined hepatocyte-mediated clearance of lipid-free and HDL-associated SAA. We prepared hepatocytes from mice injected with lipopolysaccharide or an SAA-expressing adenoviral vector. Alternatively, we incubated primary hepatocytes from SAA-deficient mice with purified SAA. We analyzed conditioned media to determine the lipidation status of endogenously produced and exogenously added SAA. Examining the migration of lipidated species, we found that SAA is lipidated and forms nascent particles that are distinct from apoA-I-containing particles and that apoA-I lipidation is unaltered when SAA is overexpressed or added to the cells, indicating that SAA is not incorporated into apoA-I-containing HDL during HDL biogenesis. Like apoA-I formation, generation of SAA-containing particles was dependent on ABCA1, but not on scavenger receptor class B type I. Hepatocytes degraded significantly more SAA than apoA-I. Taken together, our results indicate that SAA's lipidation and metabolism by the liver is independent of apoA-I and that SAA is not incorporated into HDL during HDL biogenesis.
Collapse
Affiliation(s)
- Ailing Ji
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Xuebing Wang
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | | | - Drew Jennings
- Departments of Agricultural and Medical Biotechnology, University of Kentucky, Lexington, KY
| | - Maria C de Beer
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY.,Physiology, University of Kentucky, Lexington, KY
| | - Frederick C de Beer
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY.,Internal Medicine, University of Kentucky, Lexington, KY
| | - Lisa R Tannock
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY.,Internal Medicine, University of Kentucky, Lexington, KY
| | - Nancy R Webb
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY .,Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY
| |
Collapse
|
19
|
Tong H, Zhang X, Tan L, Jin R, Huang S, Li X. Multitarget and promising role of dihydromyricetin in the treatment of metabolic diseases. Eur J Pharmacol 2019; 870:172888. [PMID: 31866404 DOI: 10.1016/j.ejphar.2019.172888] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/01/2019] [Accepted: 12/18/2019] [Indexed: 12/30/2022]
Abstract
Dihydromyricetin (DMY or DHM), also known as ampelopsin, is the main natural flavonol compound extracted from the plant Ampelopsis grossedentata (Hand. -Mazz) W.T. Wang. In recent years, accumulating studies have been conducted to explore the extensive biological functions of DMY, including antitumor, anti-inflammation, organ-protective, and metabolic regulation effects. DMY acts as a potential preventive or therapeutic agent in treating multiple diseases, such as diabetes mellitus, atherosclerosis, nonalcoholic fatty liver disease and osteoporosis. This review article summarizes the preventive and therapeutic potential of DMY in multiple metabolic diseases and the main signaling pathways in which DMY participates to offer a comprehensive understanding and guidance for future studies.
Collapse
Affiliation(s)
- Haihui Tong
- Department of Pediatrics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jie Fang Avenue, Wuhan, Hubei Province, 430022, China.
| | - Xuejun Zhang
- Department of Orthopedics, The People's Hospital of China Three Gorges University, First People's Hospital of Yichang, No. 4 Hudi Street, Yichang, Hubei Province, 443000, China.
| | - Lingfang Tan
- Department of Pediatrics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jie Fang Avenue, Wuhan, Hubei Province, 430022, China.
| | - Runming Jin
- Department of Pediatrics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jie Fang Avenue, Wuhan, Hubei Province, 430022, China.
| | - Shilong Huang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jie Fang Avenue, Wuhan, Hubei Province, 430030, China.
| | - Xin Li
- Department of Pediatrics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jie Fang Avenue, Wuhan, Hubei Province, 430022, China.
| |
Collapse
|
20
|
Maruf A, Wang Y, Yin T, Huang J, Wang N, Durkan C, Tan Y, Wu W, Wang G. Atherosclerosis Treatment with Stimuli-Responsive Nanoagents: Recent Advances and Future Perspectives. Adv Healthc Mater 2019; 8:e1900036. [PMID: 30945462 DOI: 10.1002/adhm.201900036] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/06/2019] [Indexed: 01/04/2023]
Abstract
Atherosclerosis is the root of approximately one-third of global mortalities. Nanotechnology exhibits splendid prospects to combat atherosclerosis at the molecular level by engineering smart nanoagents with versatile functionalizations. Significant advances in nanoengineering enable nanoagents to autonomously navigate in the bloodstream, escape from biological barriers, and assemble with their nanocohort at the targeted lesion. The assembly of nanoagents with endogenous and exogenous stimuli breaks down their shells, facilitates intracellular delivery, releases their cargo to kill the corrupt cells, and gives imaging reports. All these improvements pave the way toward personalized medicine for atherosclerosis. This review systematically summarizes the recent advances in stimuli-responsive nanoagents for atherosclerosis management and its progress in clinical trials.
Collapse
Affiliation(s)
- Ali Maruf
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing University Chongqing 400030 China
| | - Yi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing University Chongqing 400030 China
| | - Tieyin Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing University Chongqing 400030 China
| | - Junli Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing University Chongqing 400030 China
| | - Nan Wang
- The Nanoscience CentreUniversity of Cambridge Cambridge CB3 0FF UK
| | - Colm Durkan
- The Nanoscience CentreUniversity of Cambridge Cambridge CB3 0FF UK
| | - Youhua Tan
- Department of Biomedical EngineeringThe Hong Kong Polytechnic University Hong Kong SAR 999077 China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing University Chongqing 400030 China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing University Chongqing 400030 China
| |
Collapse
|
21
|
Xue Y, Wu Y, Wang Q, Xue L, Su Z, Zhang C. Cellular Vehicles Based on Neutrophils Enable Targeting of Atherosclerosis. Mol Pharm 2019; 16:3109-3120. [PMID: 31082253 DOI: 10.1021/acs.molpharmaceut.9b00342] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Given the multiple interactions between neutrophils (NEs) and atherosclerosis (AS), in this study, we exploited NEs as cellular vehicles loaded with cationic liposomes for actively targeting atherosclerotic sites. The cellular vehicles based on NEs possess efficient internalization of cationic liposomes and sensitive response to the chemotaxis of atherosclerotic inflammatory cells, which ultimately realize the targeted delivery of the cargos into the target cells in vitro. Moreover, these effects also translated to significant enhancement of the accumulation of NEs' cargos into the atherosclerotic plaque in vivo after administering NE vehicles to the AS animal model. Consequently, cellular vehicles based on NEs could be a novel strategy for targeted delivery of payloads into atherosclerotic plaque, which would facilitate theranostics for AS and the development of anti-AS drugs to manage the disease.
Collapse
Affiliation(s)
- Yanan Xue
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials , China Pharmaceutical University , No. 24 Tongjiaxiang , Nanjing 210009 , China
| | - Yue Wu
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials , China Pharmaceutical University , No. 24 Tongjiaxiang , Nanjing 210009 , China
| | - Qianqian Wang
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials , China Pharmaceutical University , No. 24 Tongjiaxiang , Nanjing 210009 , China
| | - Lingjing Xue
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials , China Pharmaceutical University , No. 24 Tongjiaxiang , Nanjing 210009 , China
| | - Zhigui Su
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials , China Pharmaceutical University , No. 24 Tongjiaxiang , Nanjing 210009 , China
| | - Can Zhang
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials , China Pharmaceutical University , No. 24 Tongjiaxiang , Nanjing 210009 , China
| |
Collapse
|
22
|
Chen Y, Dong J, Zhang X, Chen X, Wang L, Chen H, Ge J, Jiang XC. Evacetrapib reduces preβ-1 HDL in patients with atherosclerotic cardiovascular disease or diabetes. Atherosclerosis 2019; 285:147-152. [PMID: 31054484 DOI: 10.1016/j.atherosclerosis.2019.04.211] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/28/2019] [Accepted: 04/05/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND AND AIMS Cholesteryl ester transfer protein (CETP) inhibitor-mediated induction of HDL-cholesterol has no effect on the protection from cardiovascular disease (CVD). However, the mechanism is still unknown. Data on the effects of this class of drugs on subclasses of HDL are either limited or insufficient. In this study, we investigated the effect of evacetrapib, a CETP inhibitor, on subclasses of HDL in patients with atherosclerotic cardiovascular disease or diabetes. METHODS Baseline and 3-month post-treatment samples from atorvastatin 40 mg plus evacetrapib 130 mg (n = 70) and atorvastatin 40 mg plus placebo (n = 30) arms were used for this purpose. Four subclasses of HDL (large HDL, medium HDL, small HDL, and preβ-1 HDL) were separated according to their size and quantified by densitometry using a recently developed native polyacrylamide gel electrophoresis (PAGE) system. RESULTS Relative to placebo, while evacetrapib treatment dramatically increased large HDL and medium HDL subclasses, it significantly reduced small HDL (27%) as well as preβ-1 HDL (36%) particles. Evacetrapib treatment reduced total LDL, but also resulted in polydisperse LDL with LDL particles larger and smaller than the LDL subclasses of the placebo group. CONCLUSION Evacetrapib reduced preβ-1 HDL and small HDL in patients with ASCVD or diabetes on statin. Preβ-1 HDL and medium HDL are negatively interrelated. The results could give a clue to understand the effect of CETP inhibitors on cardiovascular outcomes.
Collapse
Affiliation(s)
- Yunqin Chen
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jibin Dong
- School of Pharmacy, Fudan University, Shanghai, China
| | - Xiaojin Zhang
- Obstetrics & Gynecology Hospital, Fudan University, Shanghai, China
| | - Xueying Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haozhu Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA; School of Pharmacy, Fudan University, Shanghai, China.
| |
Collapse
|
23
|
Jeong MJ, Kim SR, Jung UJ. Schizandrin A supplementation improves nonalcoholic fatty liver disease in mice fed a high-fat and high-cholesterol diet. Nutr Res 2019; 64:64-71. [DOI: 10.1016/j.nutres.2019.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/17/2018] [Accepted: 01/07/2019] [Indexed: 12/13/2022]
|
24
|
Zhou M, Learned RM, Rossi SJ, Tian H, DePaoli AM, Ling L. Therapeutic FGF19 promotes HDL biogenesis and transhepatic cholesterol efflux to prevent atherosclerosis. J Lipid Res 2019; 60:550-565. [PMID: 30679232 PMCID: PMC6399511 DOI: 10.1194/jlr.m089961] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/08/2019] [Indexed: 12/15/2022] Open
Abstract
Fibroblast growth factor (FGF)19, an endocrine hormone produced in the gut, acts in the liver to control bile acid synthesis. NGM282, an engineered FGF19 analog, is currently in clinical development for treating nonalcoholic steatohepatitis. However, the molecular mechanisms that integrate FGF19 with cholesterol metabolic pathways are incompletely understood. Here, we report that FGF19 and NGM282 promote HDL biogenesis and cholesterol efflux from the liver by selectively modulating LXR signaling while ameliorating hepatic steatosis. We further identify ABCA1 and FGF receptor 4 as mediators of this effect, and that administration of a HMG-CoA reductase inhibitor or a blocking antibody against proprotein convertase subtilisin/kexin type 9 abolished FGF19-associated elevations in total cholesterol, HDL cholesterol (HDL-C), and LDL cholesterol in db/db mice. Moreover, we show that a constitutively active MEK1, but not a constitutively active STAT3, mimics the effect of FGF19 and NGM282 on cholesterol change. In dyslipidemic Apoe-/- mice fed a Western diet, treatment with NGM282 dramatically reduced atherosclerotic lesion area in aortas. Administration of NGM282 to healthy volunteers for 7 days resulted in a 26% increase in HDL-C levels compared with placebo. These findings outline a previously unrecognized role for FGF19 in the homeostatic control of cholesterol and may have direct impact on the clinical development of FGF19 analogs.
Collapse
Affiliation(s)
- Mei Zhou
- NGM Biopharmaceuticals, Inc., South San Francisco, CA 94080
| | - R Marc Learned
- NGM Biopharmaceuticals, Inc., South San Francisco, CA 94080
| | | | - Hui Tian
- NGM Biopharmaceuticals, Inc., South San Francisco, CA 94080
| | - Alex M DePaoli
- NGM Biopharmaceuticals, Inc., South San Francisco, CA 94080
| | - Lei Ling
- NGM Biopharmaceuticals, Inc., South San Francisco, CA 94080
| |
Collapse
|
25
|
ABCA1 Agonist Mimetic Peptide CS-6253 Induces Microparticles Release From Different Cell Types by ABCA1-Efflux-Dependent Mechanism. Can J Cardiol 2019; 35:770-781. [PMID: 31151713 DOI: 10.1016/j.cjca.2019.02.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/18/2019] [Accepted: 02/18/2019] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Small peptides based on the C-terminal domain of apo E have recently been proposed as ATP-binding cassette transporter A1 (ABCA1) agonist with therapeutic potential. Previous work has shown that a novel synthetic peptide, CS-6253, acts synergistically with apolipoprotein A-I or alone to generate high-density lipoprotein (HDL) particles; we have also shown that cells can release microparticles (50-350 nm in apparent diameter) in an ABCA1- and apolipoprotein A-I-dependent manner. The purpose of this study was to explore the ability of a novel synthetic peptide CS-6253 to induce microparticle release from various cell lines in the process of HDL biogenesis. METHODS The effects of CS-6253 on microparticle formation through the ABCA1 transporter were examined in vitro using cell-based systems and pharmacologic manipulations. RESULTS In cell-based systems combined with fast performance liquid chromatography and nano-sight-tracking analysis, we show that ABCA1 and CS-6253 mediate and increase the production of microparticles containing cholesterol. CS-6253 in baby hamster kidney cells not expressing ABCA1 (baby hamster kidney mock cells) did not alter cholesterol removal across the plasma membrane in the absence of ABCA1 expression even at high concentrations. We report that CS-6253 is not cytotoxic. CONCLUSIONS The present study shows that CS-6253 generates cholesterol containing microparticles with size heterogeneity (100-350 nm) in an ABCA1-dependent manner. We show that microparticles contribute to cell cholesterol efflux from monocyte-macrophage cells. At high doses, CS-6253 is not able to extract cholesterol from cells not expressing ABCA1, indicating that CS-6253 requires ABCA1 cooperation for cholesterol mobilization. We conclude that CS-6253 is an ABCA1 agonist peptide that promotes cellular cholesterol efflux through HDL biogenesis and microparticle formation.
Collapse
|
26
|
Karimi-Sales E, Jeddi S, Ebrahimi-Kalan A, Alipour MR. trans-Chalcone prevents insulin resistance and hepatic inflammation and also promotes hepatic cholesterol efflux in high-fat diet-fed rats: modulation of miR-34a-, miR-451-, and miR-33a-related pathways. Food Funct 2018; 9:4292-4298. [PMID: 30039136 DOI: 10.1039/c8fo00923f] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Insulin resistance and inflammation are strongly linked to non-alcoholic fatty liver disease (NAFLD) as a feature of the metabolic syndrome. Furthermore, the role of dysregulation of miR-34a, miR-451, and miR-33a in pathogenesis and progression of NAFLD has been identified. trans-Chalcone is a simple chalcone with anti-diabetic and anti-inflammatory activities. However, to the best of our knowledge, miRNA-dependent mechanisms of these protective effects under pathologic conditions are not understood. Thus, this study, for the first time, aimed to evaluate the effects of trans-Chalcone on miR-34a, miR-451, and miR-33a signaling pathways in the liver of high-fat (HF) emulsion-fed rats. To this aim, twenty-one rats were randomly and equally divided into three groups: control, which was gavaged with 10% tween 80; HF, which was gavaged with HF emulsion and 10% tween 80; and HF + trans-Chalcone (HF + TC), which was gavaged with HF emulsion and trans-Chalcone. Then, circulating levels of glucose and insulin were measured and used for the calculation of HOMA-IR. Hepatic expression levels of miR-34a, miR-451, miR-33a, SIRT1, and ABCA1 and also protein levels of ABCA1 and IL-8 were assayed. In this study, trans-chalcone increased hepatic cholesterol efflux and prevented insulin resistance and liver inflammation in HF emulsion-fed rats. These protective effects were modulated through the down-regulation of miR-34a and its associated elevation of SIRT1, the up-regulation of miR-451 which was associated with a reduction in IL-8, and the inhibition of miR-33a which was related to the elevation of ABCA1 in the liver of HF emulsion-fed rats. Therefore, trans-Chalcone exerts its beneficial effects by targeting hepatic miR-34a-, miR-451-, and miR-33a-related pathways.
Collapse
Affiliation(s)
- Elham Karimi-Sales
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | | | | |
Collapse
|
27
|
Pioglitazone attenuates aging-related disorders in aged apolipoprotein E deficient mice. Exp Gerontol 2018; 102:101-108. [DOI: 10.1016/j.exger.2017.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/03/2017] [Accepted: 12/04/2017] [Indexed: 01/08/2023]
|
28
|
Sacks FM, Jensen MK. From High-Density Lipoprotein Cholesterol to Measurements of Function: Prospects for the Development of Tests for High-Density Lipoprotein Functionality in Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2018; 38:487-499. [PMID: 29371248 DOI: 10.1161/atvbaha.117.307025] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 01/05/2018] [Indexed: 01/13/2023]
Abstract
The evidence is strong that biological functions contained in high-density lipoproteins (HDL) are antiatherogenic. These functions may track with HDL cholesterol or apolipoprotein A1 concentration to explain the strongly inverse risk curve for cardiovascular disease. Moreover, there are harmful as well as protective HDL subspecies in regard to cardiovascular disease, which could be responsible for paradoxical responses to HDL-directed treatments. Recent metabolic studies show that apolipoprotein A1-containing HDL is secreted into the circulation as mostly spherical cholesterol ester-rich lipoproteins that span the HDL size range. Most of the flux of apolipoprotein A1 HDL into and out of the circulation occurs in these spherical cholesterol-replete particles. Discoidal cholesterol-poor HDL comprises a minority of HDL secretion. We propose that much cholesterol in reverse cholesterol transport enters and exits medium and large size HDL without changing a size category, and its flux may be estimated provisionally from holoparticle clearance of cholesterol ester-rich HDL. An accurate framework for metabolism of HDL is essential to finding steady-state biomarkers that reflect HDL function in vivo. Whereas cholesterol efflux from cells to mainly discoidal HDL, mediated by ABCA1 (ATP-binding cassette transporter ABCA1), predicts cardiovascular disease, cholesterol transfers to spherical HDL also can be measured and may be relevant to protection against atherosclerosis. We propose several investigative paths on which human HDL biology may be investigated leading to convenient biomarkers of HDL quality and function having potential not only to improve risk prediction but also to more accurately target drug treatments.
Collapse
Affiliation(s)
- Frank M Sacks
- From the Departments of Nutrition and Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA.
| | - Majken K Jensen
- From the Departments of Nutrition and Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
29
|
Tannock LR, De Beer MC, Ji A, Shridas P, Noffsinger VP, den Hartigh L, Chait A, De Beer FC, Webb NR. Serum amyloid A3 is a high density lipoprotein-associated acute-phase protein. J Lipid Res 2017; 59:339-347. [PMID: 29247043 DOI: 10.1194/jlr.m080887] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/22/2017] [Indexed: 12/20/2022] Open
Abstract
Serum amyloid A (SAA) is a family of acute-phase reactants. Plasma levels of human SAA1/SAA2 (mouse SAA1.1/2.1) can increase ≥1,000-fold during an acute-phase response. Mice, but not humans, express a third relatively understudied SAA isoform, SAA3. We investigated whether mouse SAA3 is an HDL-associated acute-phase SAA. Quantitative RT-PCR with isoform-specific primers indicated that SAA3 and SAA1.1/2.1 are induced similarly in livers (∼2,500-fold vs. ∼6,000-fold, respectively) and fat (∼400-fold vs. ∼100-fold, respectively) of lipopolysaccharide (LPS)-injected mice. In situ hybridization demonstrated that all three SAAs are produced by hepatocytes. All three SAA isoforms were detected in plasma of LPS-injected mice, although SAA3 levels were ∼20% of SAA1.1/2.1 levels. Fast protein LC analyses indicated that virtually all of SAA1.1/2.1 eluted with HDL, whereas ∼15% of SAA3 was lipid poor/free. After density gradient ultracentrifugation, isoelectric focusing demonstrated that ∼100% of plasma SAA1.1 was recovered in HDL compared with only ∼50% of SAA2.1 and ∼10% of SAA3. Thus, SAA3 appears to be more loosely associated with HDL, resulting in lipid-poor/free SAA3. We conclude that SAA3 is a major hepatic acute-phase SAA in mice that may produce systemic effects during inflammation.
Collapse
Affiliation(s)
- Lisa R Tannock
- Departments of Internal Medicine, University of Kentucky, Lexington, KY.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY.,Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY.,Veterans Affairs Lexington, University of Kentucky, Lexington, KY
| | - Maria C De Beer
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY.,Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY.,Departments of Physiology, University of Kentucky, Lexington, KY
| | - Ailing Ji
- Departments of Internal Medicine, University of Kentucky, Lexington, KY.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Preetha Shridas
- Departments of Internal Medicine, University of Kentucky, Lexington, KY.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY.,Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY
| | - Victoria P Noffsinger
- Departments of Internal Medicine, University of Kentucky, Lexington, KY.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Laura den Hartigh
- Department of Medicine University of Washington, Seattle, WA.,University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Alan Chait
- Department of Medicine University of Washington, Seattle, WA.,University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Frederick C De Beer
- Departments of Internal Medicine, University of Kentucky, Lexington, KY.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY.,Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY
| | - Nancy R Webb
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY .,Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY.,Veterans Affairs Lexington, University of Kentucky, Lexington, KY.,Departments of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY
| |
Collapse
|
30
|
Xu S, Li L, Yan J, Ye F, Shao C, Sun Z, Bao Z, Dai Z, Zhu J, Jing L, Wang Z. CML/CD36 accelerates atherosclerotic progression via inhibiting foam cell migration. Biomed Pharmacother 2017; 97:1020-1031. [PMID: 29136780 DOI: 10.1016/j.biopha.2017.11.041] [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: 07/31/2017] [Revised: 10/23/2017] [Accepted: 11/03/2017] [Indexed: 12/31/2022] Open
Abstract
Among the various complications of type 2 diabetes mellitus, atherosclerosis causes the highest disability and morbidity. A multitude of macrophage-derived foam cells are retained in atherosclerotic plaques resulting not only from recruitment of monocytes into lesions but also from a reduced rate of macrophage migration from lesions. Nε-carboxymethyl-Lysine (CML), an advanced glycation end product, is responsible for most complications of diabetes. This study was designed to investigate the mechanism of CML/CD36 accelerating atherosclerotic progression via inhibiting foam cell migration. In vivo study and in vitro study were performed. For the in vivo investigation, CML/CD36 accelerated atherosclerotic progression via promoting the accumulation of macrophage-derived foam cells in aorta and inhibited macrophage-derived foam cells in aorta migrating to the para-aorta lymph node of diabetic apoE-/- mice. For the in vitro investigation, CML/CD36 inhibited RAW264.7-derived foam cell migration through NOX-derived ROS, FAK phosphorylation, Arp2/3 complex activation and F-actin polymerization. Thus, we concluded that CML/CD36 inhibited foam cells of plaque migrating to para-aorta lymph nodes, accelerating atherosclerotic progression. The corresponding mechanism may be via free cholesterol, ROS generation, p-FAK, Arp2/3, F-actin polymerization.
Collapse
Affiliation(s)
- Suining Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Jinchuan Yan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Fei Ye
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Zhengyang Bao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Zhiyin Dai
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Jie Zhu
- Department of Cardiology, Luan Affiliated Hospital of Anhui Medical University, Anhui 237005, China.
| | - Lele Jing
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
31
|
Iqbal F, Baker WS, Khan MI, Thukuntla S, McKinney KH, Abate N, Tuvdendorj D. Current and future therapies for addressing the effects of inflammation on HDL cholesterol metabolism. Br J Pharmacol 2017; 174:3986-4006. [PMID: 28326542 PMCID: PMC5660004 DOI: 10.1111/bph.13743] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/16/2017] [Accepted: 02/02/2017] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is a major cause of morbidity and mortality worldwide. Inflammatory processes arising from metabolic abnormalities are known to precipitate the development of CVD. Several metabolic and inflammatory markers have been proposed for predicting the progression of CVD, including high density lipoprotein cholesterol (HDL-C). For ~50 years, HDL-C has been considered as the atheroprotective 'good' cholesterol because of its strong inverse association with the progression of CVD. Thus, interventions to increase the concentration of HDL-C have been successfully tested in animals; however, clinical trials were unable to confirm the cardiovascular benefits of pharmaceutical interventions aimed at increasing HDL-C levels. Based on these data, the significance of HDL-C in the prevention of CVD has been called into question. Fundamental in vitro and animal studies suggest that HDL-C functionality, rather than HDL-C concentration, is important for the CVD-preventive qualities of HDL-C. Our current review of the literature positively demonstrates the negative impact of systemic and tissue (i.e. adipose tissue) inflammation in the healthy metabolism and function of HDL-C. Our survey indicates that HDL-C may be a good marker of adipose tissue health, independently of its atheroprotective associations. We summarize the current findings on the use of anti-inflammatory drugs to either prevent HDL-C clearance or improve the function and production of HDL-C particles. It is evident that the therapeutic agents currently available may not provide the optimal strategy for altering HDL-C metabolism and function, and thus, further research is required to supplement this mechanistic approach for preventing the progression of CVD. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Fatima Iqbal
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Wendy S Baker
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Madiha I Khan
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Shwetha Thukuntla
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Kevin H McKinney
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Nicola Abate
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Demidmaa Tuvdendorj
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| |
Collapse
|
32
|
Yeh YT, Chiang AN, Hsieh SC. Chinese Olive (Canarium album L.) Fruit Extract Attenuates Metabolic Dysfunction in Diabetic Rats. Nutrients 2017; 9:nu9101123. [PMID: 29036927 PMCID: PMC5691739 DOI: 10.3390/nu9101123] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 10/11/2017] [Accepted: 10/12/2017] [Indexed: 02/06/2023] Open
Abstract
Hyperglycemia and dysregulation of lipid metabolism play a crucial role in metabolic dysfunction. The aims of present study were to evaluate the ameliorative effect of the ethyl acetate fraction of Chinese olive fruit extract (CO-EtOAc) on high-fat diet (HFD) and streptozotocin (STZ)-induced diabetic rats. CO-EtOAc, rich in gallic acid and ellagic acid, could markedly decreased the body weight and epididymal adipose mass. In addition, CO-EtOAc increased serum HDL-C levels, hepatic GSH levels, and antioxidant enzyme activities; lowered blood glucose, serum levels of total cholesterol (TC), triglycerides (TG), bile acid, and tumor necrosis factor alpha (TNFα); and reduced TC and TG in liver. We further demonstrated that CO-EtOAc mildly suppressed hepatic levels of phosphorylated IRS-1, TNF-α, and IL-6, but enhanced Akt phosphorylation. The possible mechanisms of cholesterol metabolism were assessed by determining the expression of genes involved in cholesterol transportation, biosynthesis, and degradation. It was found that CO-EtOAc not only inhibited mRNA levels of SREBP-2, HMG-CoAR, SR-B1, and CYP7A1 but also increased the expression of genes, such as ABCA1 and LDLR that governed cholesterol efflux and cholesterol uptake. Moreover, the protein expressions of ABCA1 and LDLR were also significantly increased in the liver of rats supplemented with CO-EtOAc. We suggest that Chinese olive fruit may ameliorate metabolic dysfunction in diabetic rats under HFD challenge.
Collapse
Affiliation(s)
- Yu-Te Yeh
- Institute of Food Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| | - An-Na Chiang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan.
| | - Shu-Chen Hsieh
- Institute of Food Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
33
|
Wang J, Xu P, Xie X, Li J, Zhang J, Wang J, Hong F, Li J, Zhang Y, Song Y, Zheng X, Zhai Y. DBZ (Danshensu Bingpian Zhi), a Novel Natural Compound Derivative, Attenuates Atherosclerosis in Apolipoprotein E-Deficient Mice. J Am Heart Assoc 2017; 6:e006297. [PMID: 28971954 PMCID: PMC5721843 DOI: 10.1161/jaha.117.006297] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/07/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND DBZ (Danshensu Bingpian Zhi), a synthetic derivative of a natural compound found in traditional Chinese medicine, has been reported to suppress lipopolysaccharide-induced macrophage activation and lipid accumulation in vitro. The aim of this study was to assess whether DBZ could attenuate atherosclerosis at early and advanced stages. METHODS AND RESULTS The effects of DBZ on the development of atherosclerosis were studied using apolipoprotein E-deficient (apoE-/-) mice. For early treatment, 5-week-old apoE-/- mice were fed a Western diet and treated daily by oral gavage with or without DBZ or atorvastatin for 10 weeks. For advanced treatment, 5-week-old apoE-/- mice were fed a Western diet for 10 weeks to induce atherosclerosis, and then they were randomly divided into 4 groups and subjected to the treatment of vehicle, 20 mg/kg per day DBZ, 40 mg/kg per day DBZ, or 10 mg/kg per day atorvastatin for the subsequent 10 weeks. We showed that early treatment of apoE-/- mice with DBZ markedly reduced atherosclerotic lesion formation by inhibiting inflammation and decreasing macrophage infiltration into the vessel wall. Treatment with DBZ also attenuated the progression of preestablished diet-induced atherosclerotic plaques in apoE-/- mice. In addition, we showed that DBZ may affect LXR (liver X receptor) function and that treatment of macrophages with DBZ suppressed lipopolysaccharide-stimulated cell migration and oxidized low-density lipoprotein-induced foam cell formation. CONCLUSIONS DBZ potentially has antiatherosclerotic effects that involve the inhibition of inflammation, macrophage migration, leukocyte adhesion, and foam cell formation. These results suggest that DBZ may be used as a therapeutic agent for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jing Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Pengfei Xu
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Xinni Xie
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
- State key laboratory of environmental chemistry and ecotoxicology Research Center for Eco-Environmental Science Chinese Academy of Science, Beijing, China
| | - Jiao Li
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Jun Zhang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Jialin Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
- Department of Biology Science and Technology, Baotou Teacher's College, Baotou, China
- State key laboratory of environmental chemistry and ecotoxicology Research Center for Eco-Environmental Science Chinese Academy of Science, Beijing, China
| | - Fan Hong
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Jian Li
- Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Youyi Zhang
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry and College of Life Sciences, Beijing Normal University, Beijing, China
- Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Yao Song
- Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Xiaohui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China and College of Life Sciences Northwest University, Xi'an, China
| | - Yonggong Zhai
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
34
|
Lim FT, Lim SM, Ramasamy K. Cholesterol lowering by Pediococcus acidilactici LAB4 and Lactobacillus plantarum LAB12 in adult zebrafish is associated with improved memory and involves an interplay between npc1l1 and abca1. Food Funct 2017; 8:2817-2828. [PMID: 28725889 DOI: 10.1039/c7fo00764g] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study assessed the cholesterol lowering effect of Pediococcus acidilactici LAB4 and Lactobacillus plantarum LAB12 using adult zebrafish. Animals were fed with a high cholesterol diet (HCD) with/without LAB for seven weeks. Serum and liver cholesterol was quantified using colorimetric and dye staining methods. Expressions of npc1l1 and abca1 in the liver and intestine and appa in the brain were quantified using RT-PCR. Serum and liver cholesterol was significantly lowered in LAB4- and LAB12-fed zebrafish (≤64% and ≤71%, respectively), with reduced liver cholesterol deposition. The cholesterol lowering effect was accompanied by down-regulation of npc1l1 in intestines (≤28.7%), up-regulation of abca1 in the liver (≥30.5%) and down-regulation of appa in the brain (≤24.5%). A moderately strong positive Pearson correlation (r = 0.617, p < 0.01) was found between appa and serum cholesterol. LAB-fed zebrafish exhibited improved spatial learning and memory. LAB4 and LAB12 can be potentially used in preventing hypercholesterolaemia and Alzheimer's diseases.
Collapse
Affiliation(s)
- Fei Tieng Lim
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, University Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.
| | - Siong Meng Lim
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, University Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.
| | - Kalavathy Ramasamy
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, University Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
35
|
Xu Z, Dong A, Feng Z, Li J. Interleukin-32 promotes lipid accumulation through inhibition of cholesterol efflux. Exp Ther Med 2017; 14:947-952. [PMID: 28781617 DOI: 10.3892/etm.2017.4596] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 02/14/2017] [Indexed: 12/15/2022] Open
Abstract
Interleukin-32 (IL-32) is a pro-inflammatory cytokine and its effects in various inflammatory diseases have been investigated. However, the role of IL-32 on atherosclerosis, an inflammatory disease, remains unknown. The present study examined the use of IL-32α, the most abundant transcript of IL-32, in the treatment of oxidized low-density lipoprotein (ox-LDL)-stimulated THP-1 macrophages for 24 h, which simulates a foam cell formation model. The effect of IL-32α (20, 50 and 100 ng/ml) on lipid deposition in the macrophages was analyzed using Oil Red O staining, while the cholesterol efflux on apolipoprotein A-I was also measured. The mRNA and protein expression levels of peroxisome proliferator-activated receptor γ (PPARγ), liver X receptor α (LXRα), ATP-binding cassette transporter A1 (ABCA1) and ABCG1 were quantified by reverse transcription-quantitative polymerase chain reaction and western blot analysis, respectively. The results indicated that IL-32α exposure enhanced the lipid deposition and attenuated the cholesterol efflux from ox-LDL-stimulated THP-1 macrophages in a dose-dependent manner. Furthermore, the expression levels of ABCA1, LXRα and PPARγ were dose-dependently decreased by IL-32α at the mRNA and protein levels. Addition of the PPARγ agonist 15d-PGJ2 or overexpression of PPARγ in THP-1 macrophages abrogated the IL-32α-mediated inhibition of cholesterol efflux and reversed the IL-32α-mediated downregulation of ABCA1 and LXRα. In conclusion, IL-32α enhances lipid accumulation and inhibits cholesterol efflux from ox-LDL-exposed THP-1 macrophages by regulating the PPARγ-LXRα-ABCA1 pathway.
Collapse
Affiliation(s)
- Zonglei Xu
- Department of Internal Medicine Cardiovascular Medicine, Liaocheng Development Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Aizhi Dong
- VIP Ward, Shandong Liaocheng No. 2 People's Hospital, Liaocheng, Shandong 252600, P.R. China
| | - Zerui Feng
- Department of Internal Medicine Cardiovascular Medicine, Shandong Liaocheng No. 2 People's Hospital, Liaocheng, Shandong 252600, P.R. China
| | - Jing Li
- Department of Internal Medicine Cardiovascular Medicine, Ankang City Central Hospital, Ankang, Shanxi 725000, P.R. China
| |
Collapse
|
36
|
Han XB, Li HX, Jiang YQ, Wang H, Li XS, Kou JY, Zheng YH, Liu ZN, Li H, Li J, Dou D, Wang Y, Tian Y, Yang LM. Upconversion nanoparticle-mediated photodynamic therapy induces autophagy and cholesterol efflux of macrophage-derived foam cells via ROS generation. Cell Death Dis 2017; 8:e2864. [PMID: 28594401 PMCID: PMC5520901 DOI: 10.1038/cddis.2017.242] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/15/2017] [Accepted: 04/28/2017] [Indexed: 02/05/2023]
Abstract
Macrophage-derived foam cells are a major component of atherosclerotic plaques and have an important role in the progression of atherosclerotic plaques, thus posing a great threat to human health. Photodynamic therapy (PDT) has emerged as a therapeutic strategy for atherosclerosis. Here, we investigated the effect of PDT mediated by upconversion fluorescent nanoparticles encapsulating chlorin e6 (UCNPs-Ce6) on the cholesterol efflux of THP-1 macrophage-derived foam cells and explored the possible mechanism of this effect. First, we found that PDT notably enhanced the cholesterol efflux and the induction of autophagy in both THP-1 and peritoneal macrophage-derived foam cells. The autophagy inhibitor 3-methyladenine and an ATG5 siRNA significantly attenuated PDT-induced autophagy, which subsequently suppressed the ABCA1-mediated cholesterol efflux. Furthermore, the reactive oxygen species (ROS) produced by PDT were responsible for the induction of autophagy, which could be blocked by the ROS inhibitor N-acetyl cysteine (NAC). NAC also reversed the PDT-induced suppression of p-mTOR and p-Akt. Therefore, our findings demonstrate that PDT promotes cholesterol efflux by inducing autophagy, and the autophagy was mediated in part through the ROS/PI3K/Akt/mTOR signaling pathway in THP-1 and peritoneal macrophage-derived foam cells.
Collapse
Affiliation(s)
- Xiaobo B Han
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Hongxia X Li
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Yueqing Q Jiang
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Hao Wang
- Department of Food Science and Engineering, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Xuesong S Li
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Jiayuan Y Kou
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Yinghong H Zheng
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Zhongni N Liu
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Hong Li
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Jing Li
- Department of Electron Microscopic Center, Harbin Medical University, Harbin, China
| | - Dou Dou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - You Wang
- Materials Physics and Chemistry Department, Harbin Institute of Technology, Harbin, China
| | - Ye Tian
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China.,Division of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Liming M Yang
- Department of Pathophysiology, Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| |
Collapse
|
37
|
Liu TT, Zeng Y, Tang K, Chen X, Zhang W, Xu XL. Dihydromyricetin ameliorates atherosclerosis in LDL receptor deficient mice. Atherosclerosis 2017; 262:39-50. [PMID: 28500865 DOI: 10.1016/j.atherosclerosis.2017.05.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 05/02/2017] [Accepted: 05/04/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Dihydromyricetin, the most abundant flavonoid in Ampelopsis grossedentata, exerts numerous pharmacological activities, including anti-inflammatory, antioxidant, hepatoprotective, and lipid regulatory activities; however, its protective effect against atherosclerosis remains poorly understood. The aim of the present study was to evaluate the effects of dihydromyricetin on high fat diet (HFD)-induced atherosclerosis using LDL receptor deficient (LDLr-/-) mice. METHODS Blood samples were collected for determination of serum lipid profiles, oxidized LDL (ox-LDL) and pro-inflammatory cytokines. Histology, hepatic lipid content, quantification of atherosclerosis, assessment of oxidative stress and inflammation were performed on liver and aorta samples by molecular biology methods. The effects of dihydromyricetin on ox-LDL-induced human umbilical vein endothelial cells (HUVECs) dysfunction and foam cell formation were further studied. RESULTS (1) Dihydromyricetin ameliorated hyperlipidemia, reduced serum ox-LDL, IL-6 and TNF-α levels in HFD-fed LDLr-/- mice. Moreover, (2) dihydromyricetin suppressed hepatic lipid accumulation and increased protein expressions of PPARα, LXRα and ABCA1. (3) It inhibited atherosclerotic lesion formation and favoured features of plaque stability. (4) Dihydromyricetin prevented hepatic and aortic inflammation as evidenced by the reduced IL-6 and TNF-α mRNA expression; (5) it prevented hepatic and aortic oxidative stress by normalizing activities of antioxidant enzymes in the liver and suppressing reactive oxygen species generation and NOX2 protein expression in both liver and aorta; (6) it inhibited oxLDL-induced injury, monocytes adhesion and oxidative stress in HUVECs and (7) inhibited macrophage foam cell formation and enhanced cholesterol efflux. CONCLUSIONS These findings suggest that dihydromyricetin could reduce atherosclerosis via its pleiotropic effects, including improvement of endothelial dysfunction, inhibition of macrophage foam cell formation, amelioration of lipid profiles, anti-inflammatory action and anti-oxidative effect.
Collapse
Affiliation(s)
- Ting Ting Liu
- Department of Pharmacology, Nantong University Pharmacy College, Nantong, China
| | - Yi Zeng
- Department of Pharmacology, Nantong University Pharmacy College, Nantong, China
| | - Kun Tang
- Department of Pharmacology, Nantong University Pharmacy College, Nantong, China
| | - XueMeng Chen
- Department of Pharmacology, Nantong University Pharmacy College, Nantong, China
| | - Wei Zhang
- Department of Pharmacology, Nantong University Pharmacy College, Nantong, China
| | - Xiao Le Xu
- Department of Pharmacology, Nantong University Pharmacy College, Nantong, China.
| |
Collapse
|
38
|
Li Y, Jiang B, Liang P, Tong Z, Liu M, Lv Q, Liu Y, Liu X, Tang Y, Xiao X. Nucleolin protects macrophages from oxLDL-induced foam cell formation through up-regulating ABCA1 expression. Biochem Biophys Res Commun 2017; 486:364-371. [PMID: 28315324 DOI: 10.1016/j.bbrc.2017.03.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 03/13/2017] [Indexed: 12/31/2022]
Abstract
Our recent studies have indicated that nucleolin, as a multifunctional RNA-binding protein, exerts protective effects in the myocardial cells and endothelial cells under the condition of oxidative stress. However, the function of nucleolin and its potential mechanism in macrophage-derived foam cell formation remain largely unexplored. ApoE-/- mice were fed with a high-fat diet (HFD) for 10-24 weeks. Protein expression was measured by western blotting or immunofluorescence, and gene expression at the mRNA level was detected by qRT-PCR. The level of lipid in macrophages was examined by Oil Red O staining, high-performance liquid chromatography (HPLC) and NBD-cholesterol. Actinomycin D (Act D) was used to determine the stability of ABCA1 mRNA in macrophages. The interaction of nucleolin with ABCA1 mRNA was assessed using co-immunoprecipitation (co-IP). The aortas advanced plaques demonstrated significantly lower levels of nucleolin protein compared with early plaques in ApoE-/- mice, in which the macrophage foam cells occupied main body. Nucleolin expression at the mRNA and protein levels in RAW264.7 macrophages was significantly reduced by oxidized low-density lipoprotein (oxLDL) in a dose- and time-dependent manner. Furthermore, nucleolin overexpression markedly attenuated lipid accumulation in oxLDL-challenged macrophages through increasing cholesterol efflux. In addition, nucleolin overexpression significantly increased the expression of ATP-binding cassette transporter A1 (ABCA1) at the mRNA and protein levels without affecting expressions of scavenger receptors (SR)-A, SR-B1, CD36 and ATP-binding cassette transporter G1 (ABCG1) at the mRNA level. Moreover, nucleolin overexpression increased the stability of ABCA1 mRNA in macrophages, whereas nucleolin ablation abrogated the oxLDL-induced up-regulation of ABCA1. The up-regulation of ABCA1 by nucleolin resulted from its protein-RNA interaction. Our data suggested that nucleolin inhibited foam cell formation through enhancing stability of ABCA1 mRNA and subsequently increasing cholesterol efflux.
Collapse
MESH Headings
- ATP Binding Cassette Transporter 1/genetics
- ATP Binding Cassette Transporter 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism
- Animals
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Atherosclerosis/etiology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Biological Transport/drug effects
- CD36 Antigens/genetics
- CD36 Antigens/metabolism
- Cell Differentiation
- Cell Line
- Cholesterol/metabolism
- Diet, High-Fat
- Dose-Response Relationship, Drug
- Foam Cells/drug effects
- Foam Cells/metabolism
- Foam Cells/pathology
- Gene Expression Regulation
- Hyperlipidemias/etiology
- Hyperlipidemias/genetics
- Hyperlipidemias/metabolism
- Hyperlipidemias/pathology
- Lipoproteins, LDL/pharmacology
- Macrophages/drug effects
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Mice
- Mice, Knockout
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- RNA Stability
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Scavenger Receptors, Class A/genetics
- Scavenger Receptors, Class A/metabolism
- Scavenger Receptors, Class B/genetics
- Scavenger Receptors, Class B/metabolism
- Signal Transduction
- Nucleolin
Collapse
Affiliation(s)
- Yuanbin Li
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, PR China
| | - Bimei Jiang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, PR China.
| | - Pengfei Liang
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zhongyi Tong
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, PR China
| | - Meidong Liu
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, PR China
| | - Qinglan Lv
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, PR China
| | - Yanjuan Liu
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, PR China
| | - Xuanyou Liu
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, PR China
| | - Yuting Tang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, PR China
| | - Xianzhong Xiao
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, PR China
| |
Collapse
|
39
|
Zhang M, He J, Jiang C, Zhang W, Yang Y, Wang Z, Liu J. Plaque-hyaluronidase-responsive high-density-lipoprotein-mimetic nanoparticles for multistage intimal-macrophage-targeted drug delivery and enhanced anti-atherosclerotic therapy. Int J Nanomedicine 2017; 12:533-558. [PMID: 28144137 PMCID: PMC5245982 DOI: 10.2147/ijn.s124252] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Increasing evidence has highlighted the pivotal role that intimal macrophage (iMΦ) plays in the pathophysiology of atherosclerotic plaques, which represents an attractive target for atherosclerosis treatment. In this work, to address the insufficient specificity of conventional reconstituted high-density lipoprotein (rHDL) for iMΦ and its limited cholesterol efflux ability, we designed a hyaluronan (HA)-anchored core-shell rHDL. This nanoparticle achieved efficient iMΦ-targeted drug delivery via a multistage-targeting approach, and excellent cellular cholesterol removal. It contained a biodegradable poly (lactic-co-glycolic acid) (PLGA) core within a lipid bilayer, and apolipoprotein A-I (apoA-I) absorbing on the lipid bilayer was covalently decorated with HA. The covalent HA coating with superior stability and greater shielding was favorable for not only minimizing the liver uptake but also facilitating the accumulation of nanoparticles at leaky endothelium overexpressing CD44 receptors in atherosclerotic plaques. The ultimate iMΦ homing was achieved via apoA-I after HA coating degraded by hyaluronidase (HAase) (abundant in atherosclerotic plaque). The multistage-targeting mechanism was revealed on the established injured endothelium-macrophage co-culture dynamic system. Upon treatment with HAase in vitro, the nanoparticle HA-(C)-PLGA-rHDL exhibited a greater cholesterol efflux capacity compared with conventional rHDL (2.43-fold). Better targeting efficiency toward iMΦ and attenuated liver accumulation were further proved by results from ex vivo imaging and iMΦ-specific fluorescence localization. Ultimately, HA-(C)-PLGA-rHDL loaded with simvastatin realized the most potent anti-atherogenic efficacies in model animals over other preparations. Thus, the HAase-responsive HDL-mimetic nanoparticle was shown in this study to be a promising nanocarrier for anti-atherogenic therapy, in the light of efficient iMΦ-targeted drug delivery and excellent function of mediating cellular cholesterol efflux.
Collapse
Affiliation(s)
- Mengyuan Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Jianhua He
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Cuiping Jiang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Yun Yang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Zhiyu Wang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People’s Republic of China
| |
Collapse
|
40
|
Chen R, Cheng Z, Huang J, Liu D, Wu C, Guo P, Lin W. Versicotides D–F, new cyclopeptides with lipid-lowering activities. RSC Adv 2017. [DOI: 10.1039/c7ra07940k] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Versicotides D–F, new cyclopeptides were isolated from a gorgonian-derived fungusAspergillus versicolorLZD-14-1 and showed lipid-lowering effects.
Collapse
Affiliation(s)
- Ran Chen
- State Key Laboratory of Natural and Biomimetic Drugs
- Peking University
- Beijing 100191
- China
- Pharmacology and Toxicology Research Center
| | - Zhongbin Cheng
- State Key Laboratory of Natural and Biomimetic Drugs
- Peking University
- Beijing 100191
- China
| | - Jian Huang
- State Key Laboratory of Natural and Biomimetic Drugs
- Peking University
- Beijing 100191
- China
| | - Dong Liu
- State Key Laboratory of Natural and Biomimetic Drugs
- Peking University
- Beijing 100191
- China
| | - Chongming Wu
- Pharmacology and Toxicology Research Center
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
- Beijing 100193
- PR China
| | - Peng Guo
- Pharmacology and Toxicology Research Center
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
- Beijing 100193
- PR China
| | - Wenhan Lin
- State Key Laboratory of Natural and Biomimetic Drugs
- Peking University
- Beijing 100191
- China
| |
Collapse
|
41
|
Karathanasis SK, Freeman LA, Gordon SM, Remaley AT. The Changing Face of HDL and the Best Way to Measure It. Clin Chem 2016; 63:196-210. [PMID: 27879324 DOI: 10.1373/clinchem.2016.257725] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 10/26/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND HDL cholesterol (HDL-C) is a commonly used lipid biomarker for assessing cardiovascular health. While a central focus has been placed on the role of HDL in the reverse cholesterol transport (RCT) process, our appreciation for the other cardioprotective properties of HDL continues to expand with further investigation into the structure and function of HDL and its specific subfractions. The development of novel assays is empowering the research community to assess different aspects of HDL function, which at some point may evolve into new diagnostic tests. CONTENT This review discusses our current understanding of the formation and maturation of HDL particles via RCT, as well as the newly recognized roles of HDL outside RCT. The antioxidative, antiinflammatory, antiapoptotic, antithrombotic, antiinfective, and vasoprotective effects of HDL are all discussed, as are the related methodologies for assessing these different aspects of HDL function. We elaborate on the importance of protein and lipid composition of HDL in health and disease and highlight potential new diagnostic assays based on these parameters. SUMMARY Although multiple epidemiologic studies have confirmed that HDL-C is a strong negative risk marker for cardiovascular disease, several clinical and experimental studies have yielded inconsistent results on the direct role of HDL-C as an antiatherogenic factor. As of yet, our increased understanding of HDL biology has not been translated into successful new therapies, but will undoubtedly depend on the development of alternative ways for measuring HDL besides its cholesterol content.
Collapse
Affiliation(s)
| | - Lita A Freeman
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | - Scott M Gordon
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD.
| |
Collapse
|
42
|
Pollard RD, Fulp B, Sorci-Thomas MG, Thomas MJ. High-Density Lipoprotein Biogenesis: Defining the Domains Involved in Human Apolipoprotein A-I Lipidation. Biochemistry 2016; 55:4971-81. [PMID: 27501467 DOI: 10.1021/acs.biochem.6b00347] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The first step in removing cholesterol from a cell is the ATP-binding cassette transporter 1 (ABCA1)-driven transfer of cholesterol to lipid-free or lipid-poor apolipoprotein A-I (apoA-I), which yields cholesterol-rich nascent high-density lipoprotein (nHDL) that then matures in plasma to spherical, cholesteryl ester-rich HDL. However, lipid-free apoA-I has a three-dimensional (3D) conformation that is significantly different from that of lipidated apoA-I on nHDL. By comparing the lipid-free apoA-I 3D conformation of apoA-I to that of 9-14 nm diameter nHDL, we formulated the hypothetical helical domain transitions that might drive particle formation. To test the hypothesis, ten apoA-I mutants were prepared that contained two strategically placed cysteines several of which could form intramolecular disulfide bonds and others that could not form these bonds. Mass spectrometry was used to identify amino acid sequence and intramolecular disulfide bond formation. Recombinant HDL (rHDL) formation was assessed with this group of apoA-I mutants. ABCA1-driven nHDL formation was measured in four mutants and wild-type apoA-I. The mutants contained cysteine substitutions in one of three regions: the N-terminus, amino acids 34 and 55 (E34C to S55C), central domain amino acids 104 and 162 (F104C to H162C), and the C-terminus, amino acids 200 and 233 (L200C to L233C). Mutants were studied in the locked form, with an intramolecular disulfide bond present, or unlocked form, with the cysteine thiol blocked by alkylation. Only small amounts of rHDL or nHDL were formed upon locking the central domain. We conclude that both the N- and C-terminal ends assist in the initial steps in lipid acquisition, but that opening of the central domain was essential for particle formation.
Collapse
Affiliation(s)
- Ricquita D Pollard
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine , Winston-Salem, North Carolina 27101, United States
| | - Brian Fulp
- Department of Biochemistry, Wake Forest School of Medicine , Winston-Salem, North Carolina 27101, United States
| | - Mary G Sorci-Thomas
- Departments of Medicine, Division of Endocrinology, Pharmacology and Toxicology, and Blood Research Institute, BloodCenter of Wisconsin, Medical College of Wisconsin , Milwaukee, Wisconsin 53226, United States
| | - Michael J Thomas
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| |
Collapse
|
43
|
Chen Y, Dong J, Chen X, Jiang H, Bakillah A, Zhang X, Li Z, Yin J, Liang D, Zou Y, Hussain M, Cuchel M, Rader D, Chen H, Ge J, Jiang XC. Human serum preβ1-high density lipoprotein levels are independently and negatively associated with coronary artery diseases. Nutr Metab (Lond) 2016; 13:36. [PMID: 27190545 PMCID: PMC4869297 DOI: 10.1186/s12986-016-0093-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/19/2016] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Serum preβ1-high density lipoprotein (preβ1-HDL) was defined by two-dimensional non-denaturing linear gel electrophoresis and apolipoprotein A-I immuno-blotting. Serum preβ1-HDL seems to play an important role in reverse cholesterol transport, a well-known anti-atherosclerosis process. However, there are still debatable questions for its quantification and coronary artery disease (CAD) relevance. METHODS We isolated the preβ1-HDL using a new native polyacrylamide gel electrophoresis (PAGE) system and lipid pre-staining serum. We established a two-demensional gel electrophoresis system. RESULTS We measured the preβ1-HDL in Tangier disease patients and subjects with cholesterol ester transfer protein (CETP) mutation. The preβ1-HDL is clearly separated from lipid-free apoA-I monomer and cannot be converted into other HDL particles under lecithin-cholesterol acyltransferase (LCAT) inhibition. This preβ1-HDL is a spheroidal particle with the highest apoA-1/cholesterol ratio and highest density (≥1.21 g/ml), as compared with all other HDLs. Importantly, we found that serum from subjects with Tangier disease or with cholesterol ester transfer protein (CETP) mutation have no detectible preβ1-HDL particles. We recruited a total of 102 subjects underwent diagnostic coronary angiography and measured their preβ1-HDL levels. Among them, 56 had no stenosis of coronary artery and 46 were diagnosed as CAD, which was predefined as the presence of a luminal diameter stenosis ≥50 % in at least 1 major coronary artery territory. We found that preβ1-HDL is independently and negatively associated with the severity of the coronary artery stenosis (Gensini score). CONCLUSION We established a novel and simple method for human serum preβ1-HDL quantification. We found that human lower preβ1-HDL is an independent predictor for severer coronary artery stenosis.
Collapse
Affiliation(s)
- Yunqin Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Cell Biology, SUNY Downstate Medical Center, 450 Clarkson Ave, Box 5, Brooklyn, NY 11203 USA
| | - Jibin Dong
- School of Pharmacy, Fudan University, Shanghai, China
| | - Xueying Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hui Jiang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ahmed Bakillah
- Department of Cell Biology, SUNY Downstate Medical Center, 450 Clarkson Ave, Box 5, Brooklyn, NY 11203 USA
| | - Xiaojin Zhang
- Obstetrics & Gynecology Hospital, Fudan University, Shanghai, China
| | - Zhiqiang Li
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia Yin
- Southern Medical University, Guangzhou, China
| | | | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mahmood Hussain
- Department of Cell Biology, SUNY Downstate Medical Center, 450 Clarkson Ave, Box 5, Brooklyn, NY 11203 USA
| | - Marina Cuchel
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | | | - Haozhu Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Medical Center, 450 Clarkson Ave, Box 5, Brooklyn, NY 11203 USA
| |
Collapse
|
44
|
Kim MH, de Beer MC, Wroblewski JM, Charnigo RJ, Ji A, Webb NR, de Beer FC, van der Westhuyzen DR. Impact of individual acute phase serum amyloid A isoforms on HDL metabolism in mice. J Lipid Res 2016; 57:969-79. [PMID: 27018443 DOI: 10.1194/jlr.m062174] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Indexed: 01/12/2023] Open
Abstract
The acute phase (AP) reactant serum amyloid A (SAA), an HDL apolipoprotein, exhibits pro-inflammatory activities, but its physiological function(s) are poorly understood. Functional differences between SAA1.1 and SAA2.1, the two major SAA isoforms, are unclear. Mice deficient in either isoform were used to investigate plasma isoform effects on HDL structure, composition, and apolipoprotein catabolism. Lack of either isoform did not affect the size of HDL, normally enlarged in the AP, and did not significantly change HDL composition. Plasma clearance rates of HDL apolipoproteins were determined using native HDL particles. The fractional clearance rates (FCRs) of apoA-I, apoA-II, and SAA were distinct, indicating that HDL is not cleared as intact particles. The FCRs of SAA1.1 and SAA2.1 in AP mice were similar, suggesting that the selective deposition of SAA1.1 in amyloid plaques is not associated with a difference in the rates of plasma clearance of the isoforms. Although the clearance rate of SAA was reduced in the absence of the HDL receptor, scavenger receptor class B type I (SR-BI), it remained significantly faster compared with that of apoA-I and apoA-II, indicating a relatively minor role of SR-BI in SAA's rapid clearance. These studies enhance our understanding of SAA metabolism and SAA's effects on AP-HDL composition and catabolism.
Collapse
Affiliation(s)
- Myung-Hee Kim
- Departments of Internal Medicine, University of Kentucky Medical Center, Lexington, KY 40536
| | - Maria C de Beer
- Physiology, University of Kentucky Medical Center, Lexington, KY 40536 Saha Cardiovascular Research Center, University of Kentucky Medical Center, Lexington, KY 40536
| | - Joanne M Wroblewski
- Departments of Internal Medicine, University of Kentucky Medical Center, Lexington, KY 40536 Saha Cardiovascular Research Center, University of Kentucky Medical Center, Lexington, KY 40536
| | - Richard J Charnigo
- Departments of Statistics and Biostatistics, University of Kentucky, Lexington, KY 40506
| | - Ailing Ji
- Departments of Internal Medicine, University of Kentucky Medical Center, Lexington, KY 40536 Saha Cardiovascular Research Center, University of Kentucky Medical Center, Lexington, KY 40536
| | - Nancy R Webb
- Saha Cardiovascular Research Center, University of Kentucky Medical Center, Lexington, KY 40536 Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, Lexington, KY 40536
| | - Frederick C de Beer
- Departments of Internal Medicine, University of Kentucky Medical Center, Lexington, KY 40536 Saha Cardiovascular Research Center, University of Kentucky Medical Center, Lexington, KY 40536
| | - Deneys R van der Westhuyzen
- Departments of Internal Medicine, University of Kentucky Medical Center, Lexington, KY 40536 Saha Cardiovascular Research Center, University of Kentucky Medical Center, Lexington, KY 40536 Molecular and Cellular Biochemistry, University of Kentucky Medical Center, Lexington, KY 40536
| |
Collapse
|
45
|
Wu C, Zhao Y, Chen R, Liu D, Liu M, Proksch P, Guo P, Lin W. Phenolic metabolites from mangrove-associated Penicillium pinophilum fungus with lipid-lowering effects. RSC Adv 2016. [DOI: 10.1039/c6ra00033a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Chemical examination of the mangrove-associated fungusPenicillium pinophilum(H608) resulted in isolation of 16 phenolic metabolites, including a new metabolite, namely 5′-hydroxypenicillide (1).
Collapse
Affiliation(s)
- Chongming Wu
- Pharmacology and Toxicology Research Center
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100193
| | - Yang Zhao
- State Key Laboratory of Natural and Biomimetic Drugs
- Peking University
- Beijing 100191
- P. R. China
| | - Ran Chen
- Pharmacology and Toxicology Research Center
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100193
| | - Dong Liu
- State Key Laboratory of Natural and Biomimetic Drugs
- Peking University
- Beijing 100191
- P. R. China
| | - Mingyue Liu
- Pharmacology and Toxicology Research Center
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100193
| | - Peter Proksch
- Institute für Pharmazeutische Biologie und Biotechnologie
- Heinrich-Heine-Universität Düsseldorf
- 40225 Düsseldorf
- Germany
| | - Peng Guo
- Pharmacology and Toxicology Research Center
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100193
| | - Wenhan Lin
- State Key Laboratory of Natural and Biomimetic Drugs
- Peking University
- Beijing 100191
- P. R. China
| |
Collapse
|
46
|
Mendivil CO, Furtado J, Morton AM, Wang L, Sacks FM. Novel Pathways of Apolipoprotein A-I Metabolism in High-Density Lipoprotein of Different Sizes in Humans. Arterioscler Thromb Vasc Biol 2015; 36:156-65. [PMID: 26543096 DOI: 10.1161/atvbaha.115.306138] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 10/21/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE A prevailing concept is that high-density lipoprotein (HDL) is secreted into the systemic circulation as a small mainly discoidal particle, which expands progressively and becomes spherical by uptake and esterification of cellular cholesterol and then contracts by cholesterol ester delivery to the liver, a process known as reverse cholesterol transport, thought to be impaired in people with low HDL cholesterol (HDLc). This metabolic framework has not been established in humans. APPROACH AND RESULTS We studied the metabolism of apolipoprotein A-I in 4 standard HDL sizes by endogenous isotopic labeling in 6 overweight adults with low HDLc and in 6 adults with normal body weight with high plasma HDLc. Contrary to expectation, HDL was secreted into the circulation in its entire size distribution from very small to very large similarly in both groups. Very small (prebeta) HDL comprised only 8% of total apolipoprotein A-I secretion. Each HDL subfraction circulated mostly within its secreted size range for 1 to 4 days and then was cleared. Enlargement of very small and medium to large and very large HDL and generation of very small from medium HDL were minor metabolic pathways. Prebeta HDL was cleared slower, whereas medium, large, and very large HDL were cleared faster in the low HDLc group. CONCLUSIONS A new model is proposed from these results in which HDL is metabolized in plasma mainly within several discrete, stable sizes across the common range of HDLc concentrations.
Collapse
Affiliation(s)
- Carlos O Mendivil
- From the School of Medicine, Universidad de los Andes, Bogotá, Colombia (C.O.M.); Section of Endocrinology, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia (C.O.M.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.O.M., J.F., A.M.M., L.W., F.M.S.); and Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (F.M.S.)
| | - Jeremy Furtado
- From the School of Medicine, Universidad de los Andes, Bogotá, Colombia (C.O.M.); Section of Endocrinology, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia (C.O.M.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.O.M., J.F., A.M.M., L.W., F.M.S.); and Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (F.M.S.)
| | - Allyson M Morton
- From the School of Medicine, Universidad de los Andes, Bogotá, Colombia (C.O.M.); Section of Endocrinology, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia (C.O.M.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.O.M., J.F., A.M.M., L.W., F.M.S.); and Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (F.M.S.)
| | - Liyun Wang
- From the School of Medicine, Universidad de los Andes, Bogotá, Colombia (C.O.M.); Section of Endocrinology, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia (C.O.M.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.O.M., J.F., A.M.M., L.W., F.M.S.); and Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (F.M.S.)
| | - Frank M Sacks
- From the School of Medicine, Universidad de los Andes, Bogotá, Colombia (C.O.M.); Section of Endocrinology, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia (C.O.M.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.O.M., J.F., A.M.M., L.W., F.M.S.); and Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (F.M.S.).
| |
Collapse
|
47
|
Spiromastixones Inhibit Foam Cell Formation via Regulation of Cholesterol Efflux and Uptake in RAW264.7 Macrophages. Mar Drugs 2015; 13:6352-65. [PMID: 26473890 PMCID: PMC4626694 DOI: 10.3390/md13106352] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/29/2015] [Accepted: 09/29/2015] [Indexed: 11/18/2022] Open
Abstract
Bioassay-guided evaluation shows that a deep sea-derived fungus, Spiromastix sp. MCCC 3A00308, possesses lipid-lowering activity. Chromatographic separation of a culture broth resulted in the isolation of 15 known depsidone-based analogues, labeled spiromastixones A–O (1–15). Each of these compounds was tested for its ability to inhibit oxidized low-density lipoprotein (oxLDL)-induced foam cell formation in RAW264.7 macrophages. Spiromastixones 6–8 and 12–14 significantly decreased oxLDL-induced lipid over-accumulation, reduced cell surface area, and reduced intracellular cholesterol concentration. Of these compounds, spiromastixones 6 and 14 exerted the strongest inhibitory effects. Spiromastixones 6 and 14 dramatically inhibited cholesterol uptake and stimulated cholesterol efflux to apolipoprotein A1 (ApoA1) and high-density lipoprotein (HDL) in RAW264.7 macrophages. Mechanistic investigation indicated that spiromastixones 6, 7, 12 and 14 significantly up-regulated the mRNA levels of ATP-binding cassette sub-family A1 (ABCA1) and down-regulated those of scavenger receptor CD36, while the transcription of ATP-binding cassette sub-family A1 (ABCG1) and proliferator-activated receptor gamma (PPARγ) were selectively up-regulated by 6 and 14. A transactivation reporter assay revealed that spiromastixones 6 and 14 remarkably enhanced the transcriptional activity of PPARγ. These results suggest that spiromastixones inhibit foam cell formation through upregulation of PPARγ and ABCA1/G1 and downregulation of CD36, indicating that spiromastixones 6 and 14 are promising lead compounds for further development as anti-atherogenic agents.
Collapse
|
48
|
Ebron K, Andersen CJ, Aguilar D, Blesso CN, Barona J, Dugan CE, Jones JL, Al-Sarraj T, Fernandez ML. A Larger Body Mass Index is Associated with Increased Atherogenic Dyslipidemia, Insulin Resistance, and Low-Grade Inflammation in Individuals with Metabolic Syndrome. Metab Syndr Relat Disord 2015; 13:458-64. [PMID: 26431271 DOI: 10.1089/met.2015.0053] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The consequences of increased body mass index (BMI) on the metabolic disorders associated with metabolic syndrome (MetS) have not been thoroughly examined. METHODS We analyzed data from 262 individuals, 97 men and 165 women (aged 18-70 years), classified with MetS to investigate whether variations in BMI could be associated with parameters of dyslipidemia, insulin resistance, or low-grade inflammation. We hypothesized that increases in BMI would positively correlate with the major dysregulations in metabolism that define MetS. For this purpose, individuals were separated into four subgroups based on their BMI: normal weight (<25 kg/m(2)), overweight (≥25 to <30 kg/m(2)), obese (≥30 to <40 kg/m(2)), and morbidly obese (≥40 kg/m(2)). RESULTS As expected, body weight and waist circumference increased significantly as BMI increased (P < 0.0001). Both systolic and diastolic blood pressure were lower in the normal BMI group compared with the other three BMI groups (P = 0.001). Markers of HDL metabolism were adversely impacted in elevated BMI groups, as both high-density lipoprotein cholesterol (HDL-C) and large HDL decreased as BMI increased (P = 0.01). BMI was negatively correlated with HDL-C (r = -0.193, P = 0.002), HDL size (r = (-)0.227, P = 0.002), and large HDL (r = -0.147, P = 0.037). In addition, plasma insulin was highest in subjects classified as morbidly obese (P < 0.0001). There was also a strong positive correlation between BMI and plasma insulin (r = 0.413, P < 0.0001), whereas adiponectin, a marker of insulin sensitivity, was negatively correlated with BMI (r = -0.288, P = 0.001). Finally, BMI was positively correlated with proinflammatory C-reactive protein (r = 0.312, P = 0.0001) and interleukin-6 (r = 0.238, P = 0.001). CONCLUSIONS The data from this study suggest that the physiological factors associated with increased BMI exacerbate the metabolic abnormalities characteristic of MetS.
Collapse
Affiliation(s)
- Kolin Ebron
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut
| | - Catherine J Andersen
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut.,2 Department of Biology, Fairfield University , Fairfield, Connecticut
| | - David Aguilar
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut
| | - Christopher N Blesso
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut
| | - Jacqueline Barona
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut.,3 Escuela de Microbiologia, Universidad de Antioquia , Medellín, Colombia
| | - Christine E Dugan
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut
| | - Jennifer L Jones
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut
| | - Taif Al-Sarraj
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut
| | - Maria Luz Fernandez
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut
| |
Collapse
|
49
|
Xu YY, Du F, Meng B, Xie GH, Cao J, Fan D, Yu H. Hepatic overexpression of methionine sulfoxide reductase A reduces atherosclerosis in apolipoprotein E-deficient mice. J Lipid Res 2015; 56:1891-900. [PMID: 26318157 PMCID: PMC4583078 DOI: 10.1194/jlr.m058776] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Indexed: 12/21/2022] Open
Abstract
Methionine sulfoxide reductase A (MsrA), a specific enzyme that converts methionine-S-sulfoxide to methionine, plays an important role in the regulation of protein function and the maintenance of redox homeostasis. In this study, we examined the impact of hepatic MsrA overexpression on lipid metabolism and atherosclerosis in apoE-deficient (apoE−/−) mice. In vitro study showed that in HepG2 cells, lentivirus-mediated human MsrA (hMsrA) overexpression upregulated the expression levels of several key lipoprotein-metabolism-related genes such as liver X receptor α, scavenger receptor class B type I, and ABCA1. ApoE−/− mice were intravenously injected with lentivirus to achieve high-level hMsrA expression predominantly in the liver. We found that hepatic hMsrA expression significantly reduced plasma VLDL/LDL levels, improved plasma superoxide dismutase, and paraoxonase-1 activities, and decreased plasma serum amyloid A level in apoE−/− mice fed a Western diet, by significantly altering the expression of several genes in the liver involving cholesterol selective uptake, conversion and excretion into bile, TG biosynthesis, and inflammation. Moreover, overexpression of hMsrA resulted in reduced hepatic steatosis and aortic atherosclerosis. These results suggest that hepatic MsrA may be an effective therapeutic target for ameliorating dyslipidemia and reducing atherosclerosis-related cardiovascular diseases.
Collapse
Affiliation(s)
- Yan-Yong Xu
- Department of Biochemistry and Molecular Biology Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Fen Du
- Department of Biochemistry and Molecular Biology Wuhan University School of Basic Medical Sciences, Wuhan, China Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, China Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC
| | - Bing Meng
- Department of Biochemistry and Molecular Biology Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Guang-Hui Xie
- Department of Biochemistry and Molecular Biology Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Jia Cao
- Department of Biochemistry and Molecular Biology Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Daping Fan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC
| | - Hong Yu
- Department of Biochemistry and Molecular Biology Wuhan University School of Basic Medical Sciences, Wuhan, China Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, China
| |
Collapse
|
50
|
Abstract
Cardiovascular disease is a major cause of morbidity and mortality in patients with type 2 diabetes mellitus, with a two- to fourfold increase in cardiovascular disease risk compared with non-diabetic individuals. Abnormalities in lipid metabolism that are observed in the context of type 2 diabetes are among the major factors contributing to an increased cardiovascular risk. Diabetic dyslipidaemia includes not only quantitative lipoprotein abnormalities, but also qualitative and kinetic abnormalities that, together, result in a shift towards a more atherogenic lipid profile. The primary quantitative lipoprotein abnormalities are increased triacylglycerol (triglyceride) levels and decreased HDL-cholesterol levels. Qualitative lipoprotein abnormalities include an increase in large, very low-density lipoprotein subfraction 1 (VLDL1) and small, dense LDLs, as well as increased triacylglycerol content of LDL and HDL, glycation of apolipoproteins and increased susceptibility of LDL to oxidation. The main kinetic abnormalities are increased VLDL1 production, decreased VLDL catabolism and increased HDL catabolism. In addition, even though LDL-cholesterol levels are typically normal in patients with type 2 diabetes, LDL particles show reduced turnover, which is potentially atherogenic. Although the pathophysiology of diabetic dyslipidaemia is not fully understood, the insulin resistance and relative insulin deficiency observed in patients with type 2 diabetes are likely to contribute to these lipid changes, as insulin plays an important role in regulating lipid metabolism. In addition, some adipocytokines, such as adiponectin or retinol-binding protein 4, may also contribute to the development of dyslipidaemia in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Bruno Vergès
- Service Endocrinologie, Diabétologie et Maladies Métaboliques, Hôpital du Bocage, 2 bd Maréchal de Lattre de Tassigny, 21000, Dijon, France,
| |
Collapse
|