101
|
Pathophysiology of Type 2 Diabetes Mellitus. Int J Mol Sci 2020; 21:ijms21176275. [PMID: 32872570 PMCID: PMC7503727 DOI: 10.3390/ijms21176275] [Citation(s) in RCA: 1017] [Impact Index Per Article: 254.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
Abstract
Type 2 Diabetes Mellitus (T2DM), one of the most common metabolic disorders, is caused by a combination of two primary factors: defective insulin secretion by pancreatic β-cells and the inability of insulin-sensitive tissues to respond appropriately to insulin. Because insulin release and activity are essential processes for glucose homeostasis, the molecular mechanisms involved in the synthesis and release of insulin, as well as in its detection are tightly regulated. Defects in any of the mechanisms involved in these processes can lead to a metabolic imbalance responsible for the development of the disease. This review analyzes the key aspects of T2DM, as well as the molecular mechanisms and pathways implicated in insulin metabolism leading to T2DM and insulin resistance. For that purpose, we summarize the data gathered up until now, focusing especially on insulin synthesis, insulin release, insulin sensing and on the downstream effects on individual insulin-sensitive organs. The review also covers the pathological conditions perpetuating T2DM such as nutritional factors, physical activity, gut dysbiosis and metabolic memory. Additionally, because T2DM is associated with accelerated atherosclerosis development, we review here some of the molecular mechanisms that link T2DM and insulin resistance (IR) as well as cardiovascular risk as one of the most important complications in T2DM.
Collapse
|
102
|
Wang H, Xie Y, Salvador AM, Zhang Z, Chen K, Li G, Xiao J. Exosomes: Multifaceted Messengers in Atherosclerosis. Curr Atheroscler Rep 2020; 22:57. [PMID: 32772195 DOI: 10.1007/s11883-020-00871-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW Atherosclerosis (AS) is a chronic inflammatory disease that contributes to the development of coronary artery disease, which has become a leading health burden worldwide. Though several strategies such as pharmacological treatment, exercise intervention, and surgery have been used in clinical practice, there is still no effective strategy to cure AS. Exosomes are extensively studied both as diagnostic markers as well as for therapeutic purposes due to their role in pathological processes related to AS. To elucidate the role of exosomes in AS and thus provide a new insight into AS therapy, we review recent advances concerning exosome targets and their function in mediating intercellular communication in AS, and expect to provide a reference for novel effective strategies to cure AS. RECENT FINDINGS Exosomes exert important roles in the diagnosis, development, and potential therapy of AS. For AS development, (1) activation of CD-137 in endothelial cells represses exosomal-TET2 production, causing a phenotypic switch of vascular smooth muscle cells (VSMC) and promoting plaque formation; (2) exosomal-MALTA1 derived from endothelial cells causes neutrophil extracellular traps (NETs) and M2 macrophage polarization, which aggravates AS; and (3) exosomal-miR-21-3p derived from macrophages inhibits PTEN expression and further promotes VSMC migration/proliferation, leading to AS development. For AS diagnosis, plasma exosomal-miR30e and miR-92a are considered to be potential diagnostic markers. For AS therapy, adipose mesenchymal stem cell-derived exosomes protect endothelial cells from AS aggravation, via inhibiting miR-342-5p. Exosome-mediated cross-talk between different cells within the vasculature exerts crucial roles in regulating endothelial function, proliferation and differentiation of vascular smooth muscle cells, and platelet activation as well as macrophage activation, collectively leading to the development and progression of AS. Exosomes can potentially be used as diagnostic biomarkers and constitute as a new therapeutic strategy for AS.
Collapse
Affiliation(s)
- Hongyun Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yuling Xie
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Ane M Salvador
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02214, USA
| | - Zhongrong Zhang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Kaichuan Chen
- Department of Ophthalmology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, China
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02214, USA
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China.
- School of Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
103
|
Nobeyama T, Shigyou K, Nakatsuji H, Sugiyama H, Komura N, Ando H, Hamada T, Murakami T. Control of Lipid Bilayer Phases of Cell-Sized Liposomes by Surface-Engineered Plasmonic Nanoparticles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:7741-7746. [PMID: 32502354 DOI: 10.1021/acs.langmuir.0c00049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Liquid-ordered (Lo)-phase domains, a cholesterol-rich area on lipid bilayers, have attracted significant attention recently because of their relevance to lipid rafts, the formation/collapse of which is associated with various kinds of information exchange through the plasma membrane. Here, we demonstrate that the formation/collapse of Lo-phase domains in cell-sized liposomes, that is, giant unilamellar vesicles (GUVs), can be controlled with bioactive plasmonic nanoparticles and light. The nanoparticles were prepared by surface modification of gold nanorods (AuNRs) using a cationized mutant of high-density lipoprotein (HDL), which is a natural cholesterol transporter. Upon the addition of surface-engineered AuNRs to GUVs with the mixed domains of Lo and liquid-disorder (Ld) phases, the Lo domains collapsed and solid-ordered (So)-phase domains were formed. The reverse phase transition was achieved photothermally, with the AuNRs loaded with cholesterol. During these transitions, the AuNRs appeared to be selectively localized on the less fluidic domain (Lo or So) in the phase-mixed GUVs. These results indicate that the phase transitions occur through the membrane binding of the AuNRs followed by spontaneous/photothermal transfer of cholesterol between the AuNRs and GUVs. Our strategy to develop bioactive AuNRs potentially enables spatiotemporal control of the formation/collapse of lipid rafts in living cells.
Collapse
Affiliation(s)
- Tomohiro Nobeyama
- Graduate School of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan
| | - Kazuki Shigyou
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Hirotaka Nakatsuji
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Osaka 565-0871, Japan
| | - Hiroshi Sugiyama
- Department of Chemistry, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University Institute of Advanced Study (KUIAS), Kyoto University, Yoshida-Honmachi, Sakyo-ku, Kyoto 606-8501, Japan
| | - Naoko Komura
- Institute for Glyco-core Research (iGCORE), Tokai National Higher Education and Research System, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
- Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Hiromune Ando
- Institute for Glyco-core Research (iGCORE), Tokai National Higher Education and Research System, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
- Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Tsutomu Hamada
- School of Material Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Tatsuya Murakami
- Graduate School of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University Institute of Advanced Study (KUIAS), Kyoto University, Yoshida-Honmachi, Sakyo-ku, Kyoto 606-8501, Japan
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0393, Japan
| |
Collapse
|
104
|
Silva BSDA, Lira FSD, de Freitas MC, Uzeloto JS, Dos Santos VR, Freire APCF, Bertolini GN, Gobbo LA. Traditional and elastic resistance training enhances functionality and lipid profile in the elderly. Exp Gerontol 2020; 135:110921. [PMID: 32151736 DOI: 10.1016/j.exger.2020.110921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 03/03/2020] [Accepted: 03/05/2020] [Indexed: 11/24/2022]
Affiliation(s)
- Bruna Spolador de Alencar Silva
- Skeletal Muscle Assessment Laboratory, School of Technology and Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil; Postgraduate Program in Movement Sciences, School of Technology and Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil.
| | - Fábio Santos de Lira
- Postgraduate Program in Movement Sciences, School of Technology and Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil
| | - Marcelo Conrado de Freitas
- Skeletal Muscle Assessment Laboratory, School of Technology and Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil; Postgraduate Program in Movement Sciences, School of Technology and Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil
| | - Juliana Souza Uzeloto
- Postgraduate Program in Physical Therapy, School of Technology and Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil
| | - Vanessa Ribeiro Dos Santos
- Skeletal Muscle Assessment Laboratory, School of Technology and Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil; Postgraduate Program in Movement Sciences, School of Technology and Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil
| | - Ana Paula Coelho Figueira Freire
- Postgraduate Program in Physical Therapy, School of Technology and Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil
| | - Giovana Navarro Bertolini
- Skeletal Muscle Assessment Laboratory, School of Technology and Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil; Postgraduate Program in Movement Sciences, School of Technology and Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil
| | - Luís Alberto Gobbo
- Skeletal Muscle Assessment Laboratory, School of Technology and Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil; Postgraduate Program in Movement Sciences, School of Technology and Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil; Postgraduate Program in Physical Therapy, School of Technology and Sciences, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil.
| |
Collapse
|
105
|
Paunovska K, Da Silva Sanchez A, Foster MT, Loughrey D, Blanchard EL, Islam FZ, Gan Z, Mantalaris A, Santangelo PJ, Dahlman JE. Increased PIP3 activity blocks nanoparticle mRNA delivery. SCIENCE ADVANCES 2020; 6:eaba5672. [PMID: 32743074 PMCID: PMC7375820 DOI: 10.1126/sciadv.aba5672] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 06/05/2020] [Indexed: 05/06/2023]
Abstract
The biological pathways that affect drug delivery in vivo remain poorly understood. We hypothesized that altering cell metabolism with phosphatidylinositol (3,4,5)-triphosphate (PIP3), a bioactive lipid upstream of the metabolic pathway PI3K (phosphatidylinositol 3-kinase)/AKT/ mTOR (mammalian target of rapamycin) would transiently increase protein translated by nanoparticle-delivered messenger RNA (mRNA) since these pathways increase growth and proliferation. Instead, we found that PIP3 blocked delivery of clinically-relevant lipid nanoparticles (LNPs) across multiple cell types in vitro and in vivo. PIP3-driven reductions in LNP delivery were not caused by toxicity, cell uptake, or endosomal escape. Interestingly, RNA sequencing and metabolomics analyses suggested an increase in basal metabolic rate. Higher transcriptional activity and mitochondrial expansion led us to formulate two competing hypotheses that explain the reductions in LNP-mediated mRNA delivery. First, PIP3 induced consumption of limited cellular resources, "drowning out" exogenously-delivered mRNA. Second, PIP3 triggers a catabolic response that leads to protein degradation and decreased translation.
Collapse
Affiliation(s)
| | | | - Matthew T. Foster
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | | | | - Fatima Z. Islam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Zubao Gan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Athanasios Mantalaris
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Philip J. Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | |
Collapse
|
106
|
Castaño D, Rattanasopa C, Monteiro-Cardoso VF, Corlianò M, Liu Y, Zhong S, Rusu M, Liehn EA, Singaraja RR. Lipid efflux mechanisms, relation to disease and potential therapeutic aspects. Adv Drug Deliv Rev 2020; 159:54-93. [PMID: 32423566 DOI: 10.1016/j.addr.2020.04.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
Abstract
Lipids are hydrophobic and amphiphilic molecules involved in diverse functions such as membrane structure, energy metabolism, immunity, and signaling. However, altered intra-cellular lipid levels or composition can lead to metabolic and inflammatory dysfunction, as well as lipotoxicity. Thus, intra-cellular lipid homeostasis is tightly regulated by multiple mechanisms. Since most peripheral cells do not catabolize cholesterol, efflux (extra-cellular transport) of cholesterol is vital for lipid homeostasis. Defective efflux contributes to atherosclerotic plaque development, impaired β-cell insulin secretion, and neuropathology. Of these, defective lipid efflux in macrophages in the arterial walls leading to foam cell and atherosclerotic plaque formation has been the most well studied, likely because a leading global cause of death is cardiovascular disease. Circulating high density lipoprotein particles play critical roles as acceptors of effluxed cellular lipids, suggesting their importance in disease etiology. We review here mechanisms and pathways that modulate lipid efflux, the role of lipid efflux in disease etiology, and therapeutic options aimed at modulating this critical process.
Collapse
|
107
|
Kluck GEG, Durham KK, Yoo JA, Trigatti BL. High Density Lipoprotein and Its Precursor Protein Apolipoprotein A1 as Potential Therapeutics to Prevent Anthracycline Associated Cardiotoxicity. Front Cardiovasc Med 2020; 7:65. [PMID: 32411725 PMCID: PMC7198830 DOI: 10.3389/fcvm.2020.00065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 04/06/2020] [Indexed: 01/01/2023] Open
Abstract
Cardiovascular disease and cancer are the leading causes of death in developed societies. Despite their effectiveness, many cancer therapies exhibit deleterious cardiovascular side effects such as cardiotoxicity and heart failure. The cardiotoxic effects of anthracyclines such as doxorubicin are the most well-characterized of cardiotoxic anti-cancer therapies. While other anti-neoplastic drugs also induce cardiotoxicity, often leading to heart failure, they are beyond the scope of this review. This review first summarizes the mechanisms of doxorubicin-induced cardiotoxicity. It then reviews emerging preclinical evidence that high density lipoprotein and its precursor protein apolipoprotein A1, which are known for their protective effects against ischemic cardiovascular disease, may also protect against doxorubicin-induced cardiotoxicity both directly and indirectly, when used therapeutically.
Collapse
Affiliation(s)
- George E. G. Kluck
- Department of Biochemistry and Biomedical Sciences, Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada
| | - Kristina K. Durham
- Faculty of Health Sciences, Institute of Applied Health Sciences, School of Rehabilitation Sciences, McMaster University, Hamilton, ON, Canada
| | - Jeong-Ah Yoo
- Department of Biochemistry and Biomedical Sciences, Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada
| | - Bernardo L. Trigatti
- Department of Biochemistry and Biomedical Sciences, Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada
| |
Collapse
|
108
|
An T, Zhang X, Li H, Dou L, Huang X, Man Y, Zhang X, Shen T, Li G, Li J, Tang W. GPR120 facilitates cholesterol efflux in macrophages through activation of AMPK signaling pathway. FEBS J 2020; 287:5080-5095. [PMID: 32243091 DOI: 10.1111/febs.15310] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 02/25/2020] [Accepted: 03/25/2020] [Indexed: 12/13/2022]
Abstract
Cholesterol efflux from macrophages is the initial step of reverse cholesterol transport, an important process for high-density lipoprotein-mediated atheroprotection. G protein-coupled receptor (GPR) 120, which functions as long-chain fatty acid receptor, is well known for its anti-inflammatory and insulin-sensitizing function in macrophages. However, the role of GPR120 on macrophage foam cell formation, the hallmark of atherosclerotic plaques, has not been verified. In this study, we found for the first time that stimulation of GPR120 by its agonist GW9508 elevated the expression of ATP-binding cassette transporters (ABC) A1 and ABCG1 in THP-1 macrophage-derived foam cells and Raw264.7 macrophages, and promoted ABCA1- and ABCG1-mediated cholesterol efflux and reduced cellular cholesteryl ester (CE) content as well. In addition, GPR120 activation was accompanied with the stimulation of AMPK pathway in macrophages; however, the effect of GPR120 on macrophage cholesterol efflux was largely abolished by AMPK inhibition. Moreover, the AMPK activity and the expression of ABCA1 and ABCG1 were markedly abrogated by knockdown of GPR120, or application of phospholipase C (PLC) inhibitor, calcium chelator, or CaMKK inhibitor. Because only free cholesterol can be effluxed from macrophages, we found that activation of AMPK could lead to increase both neutral CEs hydrolysis by upregulation of neutral cholesterol ester hydrolase expression and acid CEs hydrolysis by activation of ULK1. In conclusion, these results demonstrated that GPR120 facilitated ABCA1- and ABCG1-mediated cholesterol efflux through activation of PLC/Ca2+ /CaMKK/AMPK signaling pathway, which induced CE hydrolysis and elevated the expression of ABCA1 and ABCG1 in macrophages.
Collapse
Affiliation(s)
- Tong An
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.,Peking University Fifth School of Clinical Medicine (Beijing Hospital), Beijing, China
| | - Xiaoyi Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.,Peking University Fifth School of Clinical Medicine (Beijing Hospital), Beijing, China
| | - Hongxia Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Lin Dou
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiuqing Huang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yong Man
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiyue Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Tao Shen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Guoping Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Weiqing Tang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
109
|
Meilhac O, Tanaka S, Couret D. High-Density Lipoproteins Are Bug Scavengers. Biomolecules 2020; 10:biom10040598. [PMID: 32290632 PMCID: PMC7226336 DOI: 10.3390/biom10040598] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022] Open
Abstract
Lipoproteins were initially defined according to their composition (lipids and proteins) and classified according to their density (from very low- to high-density lipoproteins—HDLs). Whereas their capacity to transport hydrophobic lipids in a hydrophilic environment (plasma) is not questionable, their primitive function of cholesterol transporter could be challenged. All lipoproteins are reported to bind and potentially neutralize bacterial lipopolysaccharides (LPS); this is particularly true for HDL particles. In addition, HDL levels are drastically decreased under infectious conditions such as sepsis, suggesting a potential role in the clearance of bacterial material and, particularly, LPS. Moreover, "omics" technologies have unveiled significant changes in HDL composition in different inflammatory states, ranging from acute inflammation occurring during septic shock to low-grade inflammation associated with moderate endotoxemia such as periodontal disease or obesity. In this review, we will discuss HDL modifications associated with exposure to pathogens including bacteria, viruses and parasites, with a special focus on sepsis and the potential of HDL therapy in this context. Low-grade inflammation associated with atherosclerosis, periodontitis or metabolic syndrome may also highlight the protective role of HDLs in theses pathologies by other mechanisms than the reverse transport of cholesterol.
Collapse
Affiliation(s)
- Olivier Meilhac
- Université de la Réunion, Inserm, UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), F-97490 Sainte-Clotilde, France; (S.T.); (D.C.)
- CHU de La Réunion, Centre d’Investigations Clinique 1410, 97410 Saint-Pierre, France
- Correspondence: ; Tel.: +33-262-93-88-11
| | - Sébastien Tanaka
- Université de la Réunion, Inserm, UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), F-97490 Sainte-Clotilde, France; (S.T.); (D.C.)
- AP-HP, Service d’Anesthésie-Réanimation, CHU Bichat-Claude Bernard, 75018 Paris, France
| | - David Couret
- Université de la Réunion, Inserm, UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), F-97490 Sainte-Clotilde, France; (S.T.); (D.C.)
- CHU de La Réunion, Neurocritical Care Unit, 97410 Saint-Pierre, France
| |
Collapse
|
110
|
Fé-Gonçalves LM, Araújo JDA, Santos CHDAD, Val AL, Almeida-Val VMFD. How will farmed populations of freshwater fish deal with the extreme climate scenario in 2100? Transcriptional responses of Colossoma macropomum from two Brazilian climate regions. J Therm Biol 2020; 89:102487. [PMID: 32364997 DOI: 10.1016/j.jtherbio.2019.102487] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/29/2019] [Accepted: 12/22/2019] [Indexed: 12/27/2022]
Abstract
Tambaqui (Colossoma macropomum Cuvier, 1818) is an endemic fish of the Amazon and Orinoco basins, and it is the most economically important native species in Brazil being raised in five climatically distinct regions. In the face of current global warming, environmental variations in farm ponds represent additional challenges that may drive new adaptive regional genetic variations among broodstocks of tambaqui. In an experimental context based on the high-emission scenario of the 5th Intergovernmental Panel on Climate Change (IPCC) report, we used two farmed tambaqui populations to test this hypothesis. RNA-seq transcriptome analysis was performed in the liver of juvenile tambaqui from northern (Balbina Experimental Station, Balbina, AM) and southeastern (Brumado Fish Farming, Mogi Mirim, SP) Brazilian regions kept for 30 days in artificial environmental rooms mimicking the current and extreme climate scenarios. Three Illumina MiSeq runs produced close to 120 million 500 bp paired-end reads; 191,139 contigs were assembled with N50 = 1595. 355 genes were differentially expressed for both populations in response to the extreme scenario. After enrichment analysis, each population presented a core set of genes to cope with climate change. Northern fish induced genes related to the cellular response to stress, activation of MAPK activity, response to unfolded protein, protein metabolism and cellular response to DNA damage stimuli. Genes biologically involved in regulating cell proliferation, protein stabilisation and protein ubiquitination for degradation through the ubiquitin-proteasome system were downregulated. Genes associated with biological processes, including the cellular response to stress, MAPK cascade activation, homeostatic processes and positive regulation of immune responses were upregulated in southeastern fish. The downregulated genes were related to cytoskeleton organisation, energy metabolism, and the regulation of transcription and biological rhythms. Our findings reveal the signatures of promising candidate genes involved in the regional plasticity of each population of tambaqui in dealing with upcoming climate changes.
Collapse
Affiliation(s)
- Luciana Mara Fé-Gonçalves
- Laboratory of Ecophysiology and Molecular Evolution, Brazilian National Institute for Research of the Amazon, André Araújo Avenue, 2936, 69067-375, Petrópolis, Manaus, AM, Brazil.
| | - José Deney Alves Araújo
- Computational Systems Biology Laboratory, University of São Paulo, Professor Lúcio Martins Rodrigues Avenue, 370, 05508020, Butantã, São Paulo, SP, Brazil
| | - Carlos Henrique Dos Anjos Dos Santos
- Laboratory of Ecophysiology and Molecular Evolution, Brazilian National Institute for Research of the Amazon, André Araújo Avenue, 2936, 69067-375, Petrópolis, Manaus, AM, Brazil
| | - Adalberto Luis Val
- Laboratory of Ecophysiology and Molecular Evolution, Brazilian National Institute for Research of the Amazon, André Araújo Avenue, 2936, 69067-375, Petrópolis, Manaus, AM, Brazil
| | - Vera Maria Fonseca de Almeida-Val
- Laboratory of Ecophysiology and Molecular Evolution, Brazilian National Institute for Research of the Amazon, André Araújo Avenue, 2936, 69067-375, Petrópolis, Manaus, AM, Brazil
| |
Collapse
|
111
|
Lorenzatti AJ, Toth PP. New Perspectives on Atherogenic Dyslipidaemia and Cardiovascular Disease. Eur Cardiol 2020; 15:1-9. [PMID: 32180834 PMCID: PMC7066832 DOI: 10.15420/ecr.2019.06] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022] Open
Abstract
Over the past few decades, atherogenic dyslipidaemia has become one of the most common phenotypic presentations of lipid abnormalities, being strongly and unequivocally associated with an increased risk of cardiovascular (CV) disease. Despite the excellent results achieved from statin and non-statin management of LDL cholesterol and CV events prevention, there still remains a significant residual risk, associated with the prevalence of non-LDL cholesterol lipid patterns characterised by elevated triglyceride levels, low HDL cholesterol, a preponderance of small and dense LDL particles, accumulation of remnant lipoproteins and postprandial hyperlipidaemia. These qualitative and quantitative lipid modifications are largely associated with insulin resistance, type 2 diabetes and obesity, the prevalence of which has grown to epidemic proportions throughout the world. In this review, we analyse the pathophysiology of this particular dyslipidaemia, its relationship with the development of atherosclerotic CV disease and, finally, briefly describe the therapeutic approaches, including changes in lifestyle and current pharmacological interventions to manage these lipid alterations aimed at preventing CV events.
Collapse
Affiliation(s)
- Alberto J Lorenzatti
- DAMIC Medical Institute, Rusculleda Foundation for Research, Cordoba, Argentina.,Department of Cardiology, Cordoba Hospital, Cordoba, Argentina
| | - Peter P Toth
- CGH Medical Center, Sterling, IL, US.,Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, US
| |
Collapse
|
112
|
Barros D, García-Río F. Obstructive sleep apnea and dyslipidemia: from animal models to clinical evidence. Sleep 2020; 42:5204276. [PMID: 30476296 DOI: 10.1093/sleep/zsy236] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 10/02/2018] [Accepted: 11/22/2018] [Indexed: 01/11/2023] Open
Abstract
Lipid metabolism deregulation constitutes the pathogenic basis for the development of atherosclerosis and justifies a high incidence of cardiovascular-related morbidity and mortality. Some data suggest that dyslipidemia may be associated with sleep-disordered breathing, mainly obstructive sleep apnea (OSA), due to alterations in fundamental biochemical processes, such as intermittent hypoxia (IH). The aim of this systematic review was to identify and critically evaluate the current evidence supporting the existence of a possible relationship between OSA and alterations in lipid metabolism. Much evidence shows that, during the fasting state, OSA and IH increase lipid delivery from the adipose tissue to the liver through an up-regulation of the sterol regulatory element-binding protein-1 and stearoyl-CoA desaturase-1, increasing the synthesis of cholesterol esters and triglycerides. In the postprandial state, lipoprotein clearance is delayed due to lower lipoprotein lipase activity, probably secondary to IH-up-regulation of angiopoietin-like protein 4 and decreased activity of the peroxisome proliferator-activated receptor alpha. Moreover, oxidative stress can generate dysfunctional oxidized lipids and reduce the capacity of high-density lipoproteins (HDL) to prevent low-density lipoprotein (LDL) oxidation. In the clinical field, several observational studies and a meta-regression analysis support the existence of a link between OSA and dyslipidemia. Although there is evidence of improved lipid profile after apnea-hypopnea suppression with continuous positive airway pressure (CPAP), the majority of the data come from observational studies. In contrast, randomized controlled trials evaluating the effects of CPAP on lipid metabolism present inconclusive results and two meta-analyses provide contradictory evidence.
Collapse
Affiliation(s)
- David Barros
- Servicio de Neumología, Hospital Montecelo, Pontevedra, Spain
| | - Francisco García-Río
- Servicio de Neumología, Hospital Universitario La Paz-IdiPAZ, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
113
|
Zamanian-Daryoush M, Gogonea V, DiDonato AJ, Buffa JA, Choucair I, Levison BS, Hughes RA, Ellington AD, Huang Y, Li XS, DiDonato JA, Hazen SL. Site-specific 5-hydroxytryptophan incorporation into apolipoprotein A-I impairs cholesterol efflux activity and high-density lipoprotein biogenesis. J Biol Chem 2020; 295:4836-4848. [PMID: 32098873 DOI: 10.1074/jbc.ra119.012092] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/13/2020] [Indexed: 12/20/2022] Open
Abstract
Apolipoprotein A-I (apoA-I) is the major protein constituent of high-density lipoprotein (HDL) and a target of myeloperoxidase-dependent oxidation in the artery wall. In atherosclerotic lesions, apoA-I exhibits marked oxidative modifications at multiple sites, including Trp72 Site-specific mutagenesis studies have suggested, but have not conclusively shown, that oxidative modification of Trp72 of apoA-I impairs many atheroprotective properties of this lipoprotein. Herein, we used genetic code expansion technology with an engineered Saccharomyces cerevisiae tryptophanyl tRNA-synthetase (Trp-RS):suppressor tRNA pair to insert the noncanonical amino acid 5-hydroxytryptophan (5-OHTrp) at position 72 in recombinant human apoA-I and confirmed site-specific incorporation utilizing MS. In functional characterization studies, 5-OHTrp72 apoA-I (compared with WT apoA-I) exhibited reduced ABC subfamily A member 1 (ABCA1)-dependent cholesterol acceptor activity in vitro (41.73 ± 6.57% inhibition; p < 0.01). Additionally, 5-OHTrp72 apoA-I displayed increased activation and stabilization of paraoxonase 1 (PON1) activity (μmol/min/mg) when compared with WT apoA-I and comparable PON1 activation/stabilization compared with reconstituted HDL (WT apoA-I, 1.92 ± 0.04; 5-OHTrp72 apoA-I, 2.35 ± 0.0; and HDL, 2.33 ± 0.1; p < 0.001, p < 0.001, and p < 0.001, respectively). Following injection into apoA-I-deficient mice, 5-OHTrp72 apoA-I reached plasma levels comparable with those of native apoA-I yet exhibited significantly reduced (48%; p < 0.01) lipidation and evidence of HDL biogenesis. Collectively, these findings unequivocally reveal that site-specific oxidative modification of apoA-I via 5-OHTrp at Trp72 impairs cholesterol efflux and the rate-limiting step of HDL biogenesis both in vitro and in vivo.
Collapse
Affiliation(s)
- Maryam Zamanian-Daryoush
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195.,Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, Ohio 44195
| | - Valentin Gogonea
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195.,Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, Ohio 44195.,Department of Chemistry, Cleveland State University, Cleveland, Ohio 44115
| | - Anthony J DiDonato
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195.,Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, Ohio 44195
| | - Jennifer A Buffa
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195.,Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, Ohio 44195
| | - Ibrahim Choucair
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195.,Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, Ohio 44195.,Department of Chemistry, Cleveland State University, Cleveland, Ohio 44115
| | - Bruce S Levison
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Randall A Hughes
- United States Army Research Laboratory South, University of Texas, Austin, Texas 78712
| | - Andrew D Ellington
- Center for Systems and Synthetic Biology, University of Texas, Austin, Texas 78712
| | - Ying Huang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195.,Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, Ohio 44195
| | - Xinmin S Li
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195.,Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, Ohio 44195
| | - Joseph A DiDonato
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195.,Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, Ohio 44195
| | - Stanley L Hazen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195 .,Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, Ohio 44195.,Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|
114
|
Higgins V, Adeli K. Postprandial dyslipidemia in insulin resistant states in adolescent populations. J Biomed Res 2020; 34:328-342. [PMID: 32934193 PMCID: PMC7540238 DOI: 10.7555/jbr.34.20190094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
Obesity and the metabolic syndrome are becoming increasingly prevalent not only in adults, but also in adolescents. The metabolic syndrome, a complex cluster of metabolic abnormalities, increases one's risk of developing type 2 diabetes and cardiovascular disease (CVD). Dyslipidemia, a key component of the metabolic syndrome, is highly associated with insulin resistance and contributes to increased CVD risk. Dyslipidemia has traditionally been assessed using a fasting lipid profile [i.e. fasting triglycerides, total cholesterol, low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C)]. However, the postprandial state predominates over the course of a day and non-fasting triglycerides independently predict CVD risk. In insulin resistant states, the intestine overproduces triglyceride-rich lipoprotein (TRL) particles, termed chylomicrons (CMs), following ingestion of a fat-containing meal, as well as in the fasting state. Along with elevated hepatic TRLs (i.e. very-low density lipoproteins), CMs contribute to remnant lipoprotein accumulation, small dense LDL particles, and reduced HDL-C, which collectively increase CVD risk. Given the early genesis of atherosclerosis and physiological metabolic changes during adolescence, studying postprandial dyslipidemia in the adolescent population is an important area of study. Postprandial dyslipidemia in the pediatric population poses a significant public health concern, warranting a better understanding of its pathogenesis and association with insulin resistance and CVD. This review discusses the metabolic syndrome, focusing on the link between insulin resistance, postprandial dyslipidemia, and CVD risk. Furthermore, the clinical significance and functional assessment of postprandial dyslipidemia, specifically in the adolescent population, is discussed in more detail.
Collapse
Affiliation(s)
- Victoria Higgins
- Molecular Medicine and Pediatric Laboratory Medicine, Research Institute, The Hospital for Sick Children
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Khosrow Adeli
- Molecular Medicine and Pediatric Laboratory Medicine, Research Institute, The Hospital for Sick Children
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
115
|
Xu Y, Li F, Zhao X, Tan C, Wang B, Chen Y, Cao J, Wu D, Yu H. Methionine sulfoxide reductase A attenuates atherosclerosis via repairing dysfunctional HDL in scavenger receptor class B type I deficient mice. FASEB J 2020; 34:3805-3819. [PMID: 31975555 DOI: 10.1096/fj.201902429r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 01/10/2023]
Abstract
High-density lipoprotein (HDL), a well-known atheroprotective factor, can be converted to proatherogenic particles in chronic inflammation. HDL-targeted therapeutic strategy for atherosclerotic cardiovascular disease (CVD) is currently under development. This study aims to assess the role of methionine sulfoxide reductase A (MsrA) in abnormal HDL and its related disorders in scavenger receptor class B type I deficient (SR-BI-/- ) mice. First, we demonstrated that MsrA overexpression attenuated ROS level and inflammation in HepG2 cells. For the in vivo study, SR-BI-/- mice were intravenously injected with lentivirus to achieve hepatic MsrA overexpression. High-level hepatic MsrA significantly reduced the plasma free cholesterol contents, improved HDL functional proteins apolipoprotein A-I (apoAI), apoE, paraoxonase1 (PON1), and lecithin:cholesterol acyltransferase (LCAT), while decreased the pro-inflammatory property of dysfunctional HDL, contributing to reduced atherosclerosis and hepatic steatosis in Western diet-fed mice. Furthermore, the study revealed that hepatic MsrA altered the expression of several genes controlling HDL biogenesis, cholesterol esterification, cholesterol uptake mediated by low-density lipoprotein receptor (LDLR) and biliary excretion, as well as suppressed nuclear factor κB (NF-κB) signaling pathway, which largely relied on liver X receptor alpha (LXRα)-upregulation. These results provide original evidence that MsrA may be a promising target for the therapy of dysfunctional HDL-related CVD.
Collapse
Affiliation(s)
- Yanyong Xu
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Feifei Li
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Xiaojie Zhao
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Chenkun Tan
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Binyi Wang
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Yiyong Chen
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Jia Cao
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Dongfang Wu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hong Yu
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, China
| |
Collapse
|
116
|
Wang HH, Liu M, Portincasa P, Wang DQH. Recent Advances in the Critical Role of the Sterol Efflux Transporters ABCG5/G8 in Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:105-136. [PMID: 32705597 PMCID: PMC8118135 DOI: 10.1007/978-981-15-6082-8_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease is characterized by lipid accumulation, inflammatory response, cell death, and fibrosis in the arterial wall and is the leading cause of morbidity and mortality worldwide. Cholesterol gallstone disease is caused by complex genetic and environmental factors and is one of the most prevalent and costly digestive diseases in the USA and Europe. Although sitosterolemia is a rare inherited lipid storage disease, its genetic studies led to identification of the sterol efflux transporters ABCG5/G8 that are located on chromosome 2p21 in humans and chromosome 17 in mice. Human and animal studies have clearly demonstrated that ABCG5/G8 play a critical role in regulating hepatic secretion and intestinal absorption of cholesterol and plant sterols. Sitosterolemia is caused by a mutation in either the ABCG5 or the ABCG8 gene alone, but not in both simultaneously. Polymorphisms in the ABCG5/G8 genes are associated with abnormal plasma cholesterol metabolism and may play a key role in the genetic determination of plasma cholesterol concentrations. Moreover, ABCG5/G8 is a new gallstone gene, LITH9. Gallstone-associated variants in ABCG5/G8 are involved in the pathogenesis of cholesterol gallstones in European, Asian, and South American populations. In this chapter, we summarize the latest advances in the critical role of the sterol efflux transporters ABCG5/G8 in regulating hepatic secretion of biliary cholesterol, intestinal absorption of cholesterol and plant sterols, the classical reverse cholesterol transport, and the newly established transintestinal cholesterol excretion, as well as in the pathogenesis and pathophysiology of ABCG5/G8-related metabolic diseases such as sitosterolemia, cardiovascular disease, and cholesterol gallstone disease.
Collapse
Affiliation(s)
- Helen H Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Min Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Piero Portincasa
- Department of Biomedical Sciences and Human Oncology, Clinica Medica "A. Murri", University of Bari Medical School, Bari, Italy
| | - David Q-H Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
117
|
The effects of statins with a high hepatoselectivity rank on the extra-hepatic tissues; New functions for statins. Pharmacol Res 2019; 152:104621. [PMID: 31891788 DOI: 10.1016/j.phrs.2019.104621] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 12/26/2019] [Indexed: 12/14/2022]
Abstract
Statins, as the most common treatment for hyperlipidemia, exert effects beyond their lipid-lowering role which are known as pleiotropic effects. These effects are mainly due to the inhibition of isoprenoids synthesis and consequently blocking prenylation of proteins involved in the cellular signaling pathways regulating cell development, growth, and apoptosis. Statins target cholesterol synthesis in the liver as the major source of cholesterol in the body and so reduce whole-body cholesterol. The reduced level of cholesterol forces other organs to an adaptive homeostatic reaction to increase their cholesterol synthesis capacity, however, this only occurs when statins have unremarkable access to the extra-hepatic tissues. In order to reduce the adverse effects of statin on the skeletal muscle, most recent efforts have been towards formulating new statins with the highest level of hepatoselectivity rank and the least level of access to the extra-hepatic tissues; however, the inaccessibility of statins for the extra-hepatic tissues may induce several biological reactions. In this review, we aim to evaluate the effects of statins on the extra-hepatic tissues when statins have unremarkable access to these tissues.
Collapse
|
118
|
Singh RK, Lund FW, Haka AS, Maxfield FR. High-density lipoprotein or cyclodextrin extraction of cholesterol from aggregated LDL reduces foam cell formation. J Cell Sci 2019; 132:jcs.237271. [PMID: 31719160 DOI: 10.1242/jcs.237271] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/05/2019] [Indexed: 01/29/2023] Open
Abstract
Low-density lipoprotein (LDL) deposition, aggregation and retention in the endothelial sub-intima are critical initiating events during atherosclerosis. Macrophages digest aggregated LDL (agLDL) through a process called exophagy. High-density lipoprotein (HDL) plays an atheroprotective role, but studies attempting to exploit it therapeutically have been unsuccessful, highlighting gaps in our current understanding of HDL function. Here, we characterized the role of HDL during exophagy of agLDL. We find that atherosclerotic plaque macrophages contact agLDL and form an extracellular digestive compartment similar to that observed in vitro During macrophage catabolism of agLDL in vitro, levels of free cholesterol in the agLDL are increased. HDL can extract free cholesterol directly from this agLDL and inhibit macrophage foam cell formation. Cholesterol-balanced hydroxypropyl-β-cyclodextrin similarly reduced macrophage cholesterol uptake and foam cell formation. Finally, we show that HDL can directly extract free cholesterol, but not cholesterol esters, from agLDL in the absence of cells. Together, these results suggest that the actions of HDL can directly extract free cholesterol from agLDL during catabolism, and provide a new context in which to view the complex relationship between HDL and atherosclerosis.
Collapse
Affiliation(s)
- Rajesh K Singh
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Frederik W Lund
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Abigail S Haka
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Frederick R Maxfield
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
119
|
Karan S, Mohapatra A, Sahoo PK, Garg LC, Dixit A. Structural-functional characterization of recombinant Apolipoprotein A-I fromLabeo rohitademonstrates heat-resistant antimicrobial activity. Appl Microbiol Biotechnol 2019; 104:145-159. [DOI: 10.1007/s00253-019-10204-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/09/2019] [Accepted: 10/19/2019] [Indexed: 12/20/2022]
|
120
|
Kim M, Choi S, Choi SH, Shin SH, Kim SK, Shim YS, Jeon YH. Metabolic syndrome and lung function in Korean children and adolescents: A cross-sectional study. Sci Rep 2019; 9:15646. [PMID: 31666559 PMCID: PMC6821710 DOI: 10.1038/s41598-019-51968-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/10/2019] [Indexed: 12/20/2022] Open
Abstract
This study aimed to investigate whether obesity and metabolic syndrome (MetS) are associated with pulmonary function in Korean children and adolescents. Data from the 2009–2011 Korea National Health and Nutrition Examination Survey which is cross-sectional, nationwide, and representative survey were used. Adjusted regression analysis was performed to evaluate the association of obesity and MetS with lung function in children and adolescents. A total of 763 children and adolescents aged 10–18 years were evaluated. We found no significant difference in FEV1% predicted, FVC% predicted, and FEV1/FVC ratio among the obesity groups. Subjects with MetS showed a significantly lower FEV1 predicted (91.54 ± 0.74% vs 94.64 ± 0.73%, P = 0.004), lower FVC% predicted (91.86 ± 0.63% vs 95.20 ± 0.63%, P < 0.001), and lower FEV1/FVC ratio (76.76 ± 0.43% vs 80.13 ± 0.43%, P < 0.001) than those without MetS. Elevated waist circumference (WC), systolic blood pressure, fasting glucose, and lower high-density lipoprotein cholesterol (HDL-C) were independently associated with lower FEV1/FVC ratio (all P < 0.05, respectively). Among MetS components, increased WC was the most important factor influencing lower FEV1/FVC ratio. In conclusion, lung function in MetS patients was significantly lower, and the MetS component was independently associated.
Collapse
Affiliation(s)
- Minji Kim
- Department of Pediatrics, Hallym University, Dongtan Sacred Heart Hospital, Hwaseong, Gyeonggi-do, Korea.,Allergy and Clinical Immunology Research center, Hallym University College of Medicine, Chuncheon, Korea
| | - Seoheui Choi
- Department of Pediatrics, Hallym University, Dongtan Sacred Heart Hospital, Hwaseong, Gyeonggi-do, Korea
| | - Soo-Han Choi
- Department of Pediatrics, Hallym University, Dongtan Sacred Heart Hospital, Hwaseong, Gyeonggi-do, Korea
| | - Seon-Hee Shin
- Department of Pediatrics, Hallym University, Dongtan Sacred Heart Hospital, Hwaseong, Gyeonggi-do, Korea
| | - Sung Koo Kim
- Department of Pediatrics, Hallym University, Dongtan Sacred Heart Hospital, Hwaseong, Gyeonggi-do, Korea
| | - Young Suk Shim
- Department of Pediatrics, Hallym University, Dongtan Sacred Heart Hospital, Hwaseong, Gyeonggi-do, Korea.
| | - You Hoon Jeon
- Department of Pediatrics, Hallym University, Dongtan Sacred Heart Hospital, Hwaseong, Gyeonggi-do, Korea. .,Allergy and Clinical Immunology Research center, Hallym University College of Medicine, Chuncheon, Korea.
| |
Collapse
|
121
|
Gao JH, He LH, Yu XH, Zhao ZW, Wang G, Zou J, Wen FJ, Zhou L, Wan XJ, Zhang DW, Tang CK. CXCL12 promotes atherosclerosis by downregulating ABCA1 expression via the CXCR4/GSK3β/β-catenin T120/TCF21 pathway. J Lipid Res 2019; 60:2020-2033. [PMID: 31662443 DOI: 10.1194/jlr.ra119000100] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 10/22/2019] [Indexed: 12/13/2022] Open
Abstract
CXC chemokine ligand 12 (CXCL12) is a member of the CXC chemokine family and mainly acts on cell chemotaxis. CXCL12 also elicits a proatherogenic role, but the molecular mechanisms have not been fully defined yet. We aimed to reveal if and how CXCL12 promoted atherosclerosis via regulating lipid metabolism. In vitro, our data showed that CXCL12 could reduce ABCA1 expression, and it mediated cholesterol efflux from THP-1-derived macrophages to apoA-I. Data from the luciferase reporter gene and chromatin immunoprecipitation assays revealed that transcription factor 21 (TCF21) stimulated the transcription of ABCA1 via binding to its promoter region, which was repressed by CXCL12. We found that CXCL12 increased the levels of phosphorylated glycogen synthase kinase 3β (GSK3β) and the phosphorylation of β-catenin at the Thr120 position. Inactivation of GSK3β or β-catenin increased the expression of TCF21 and ABCA1. Further, knockdown or inhibition of CXC chemokine receptor 4 (CXCR4) blocked the effects of CXCL12 on TCF21 and ABCA1 expression and the phosphorylation of GSK3β and β-catenin. In vivo, the overexpression of CXCL12 in Apoe-/- mice via lentivirus enlarged the atherosclerotic lesion area and increased macrophage infiltration in atherosclerotic plaques. We further found that the overexpression of CXCL12 reduced the efficiency of reverse cholesterol transport and plasma HDL-C levels, decreased ABCA1 expression in the aorta and mouse peritoneal macrophages (MPMs), and suppressed cholesterol efflux from MPMs to apoA-I in Apoe-/- mice. Collectively, these findings suggest that CXCL12 interacts with CXCR4 and then activates the GSK-3β/β-cateninT120/TCF21 signaling pathway to inhibit ABCA1-dependent cholesterol efflux from macrophages and aggravate atherosclerosis. Targeting CXCL12 may be a novel and promising strategy for the prevention and treatment of atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Jia-Hui Gao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Lin-Hao He
- School of Pharmacy and Life Science College, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Xiao-Hua Yu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Zhen-Wang Zhao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Gang Wang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Jin Zou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Feng-Jiao Wen
- School of Pharmacy and Life Science College, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Li Zhou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Xiang-Jun Wan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| |
Collapse
|
122
|
Norring-Agerskov D, Madsen CM, Bathum L, Pedersen OB, Lauritzen JB, Jørgensen NR, Jørgensen HL. History of cardiovascular disease and cardiovascular biomarkers are associated with 30-day mortality in patients with hip fracture. Osteoporos Int 2019; 30:1767-1778. [PMID: 31278472 DOI: 10.1007/s00198-019-05056-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/12/2019] [Indexed: 11/30/2022]
Abstract
UNLABELLED Hip fractures are associated with increased mortality and it is important to identify risk factors. This study demonstrates that preexisting cardiovascular disease as well as cardiovascular biomarkers that are associated with increased 30-day mortality. These findings can be used to identify high-risk patients who might benefit from specialized care. INTRODUCTION This study investigates the association between cardiovascular disease (CVD), cardiovascular biomarkers, and 30-day mortality following a hip fracture. METHODS The Danish National Patient Registry was used to investigate the association between CVD and mortality following hip fracture in a nationwide population-based cohort study. In a subset of the included patients (n = 355), blood samples were available from a local biobank. These samples were used for analyzing the association between specific biochemical markers and mortality. The primary outcome was 30-day mortality. RESULTS A total of 113,211 patients were included in the population-based cohort study. Among these, heart failure was present in 9.4%, ischemic heart disease in 15.9%, and ischemic stroke in 12.0%. Within 30 days after the hip fracture, 11,488 patients died, resulting in an overall 30-day mortality of 10.1%. The 30-day mortality was significantly increased in individuals with preexisting CVD with multivariably adjusted odds ratios of 1.69 (95% confidence interval, 1.60-1.78) for heart failure, 1.23 (1.17-1.29) for ischemic heart disease, and 1.06 (1.00-1.12) for ischemic stroke. In the local database including 355 patients, 41 (11.5%) died within 30 days. The multivariably adjusted odds ratio for 30-day mortality increased with increasing NT-proBNP (2.36 [1.53-3.64] per quartile) and decreased with increasing HDL cholesterol (0.58 [0.41-0.82] per quartile). On this basis, we established a model for predicting the probability of death based on the biochemical markers. CONCLUSION Preexisting CVD was associated with increased 30-day mortality after a hip fracture. Furthermore, high levels of NT-proBNP and low levels of HDL cholesterol were associated with increased 30-day mortality.
Collapse
Affiliation(s)
- D Norring-Agerskov
- Department of Clinical Biochemistry, Hvidovre Hospital, Kettegård Alle 30, 2650, Hvidovre, Denmark.
- Open Patient Data Explorative Network, University of Southern Denmark and Odense University Hospital, J.B. Winsløws Vej 9A, 5000, Odense C, Denmark.
| | - C M Madsen
- Department of Clinical Biochemistry, Herlev og Gentofte Hospital, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - L Bathum
- Department of Clinical Biochemistry, Hvidovre Hospital, Kettegård Alle 30, 2650, Hvidovre, Denmark
| | - O B Pedersen
- Department of Clinical Immunology, Næstved Sygehus, Ringstedgade 61, 4700, Næstved, Denmark
| | - J B Lauritzen
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
- Department of Orthopedic Surgery, Bispebjerg Hospital, Bispebjerg Bakke 23, 2400, Copenhagen NV, Denmark
| | - N R Jørgensen
- Open Patient Data Explorative Network, University of Southern Denmark and Odense University Hospital, J.B. Winsløws Vej 9A, 5000, Odense C, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, Valdemar Hansens Vej 13, 2600, Glostrup, Denmark
| | - H L Jørgensen
- Department of Clinical Biochemistry, Hvidovre Hospital, Kettegård Alle 30, 2650, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| |
Collapse
|
123
|
Agu CE, Uchendu IK, Nsonwu AC, Okwuosa CN, Achukwu PU. Prevalence and associated risk factors of peripheral artery disease in virologically suppressed HIV-infected individuals on antiretroviral therapy in Kwara state, Nigeria: a cross sectional study. BMC Public Health 2019; 19:1143. [PMID: 31429736 PMCID: PMC6700806 DOI: 10.1186/s12889-019-7496-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 08/14/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The association between HIV and cardiovascular disease (CVD) has been reported in several studies. However, there is paucity of information on the prevalence of subclinical disease as well as its associated risk factors in sub-Saharan African population. The aim of this study was to determine the prevalence and associated risk factors of peripheral artery disease (PAD) among virologically suppressed HIV-infected participants in Kwara State, Nigeria. METHODS This study was conducted between July 2018 and December 2018. A total of 150 HIV-infected participants aged between 20 and 55 years and 50 HIV non-infected age-matched controls were randomly recruited in the study. Sociodemographic, anthropometric and clinical data were collected using a well-structured questionnaire. Ankle brachial index (ABI) was measured, PAD was defined as ABI of < 0.9. Cryopreserved serum was used to evaluate lipid profile parameters. Student's t-test and Chi-square were used to compare continuous and categorical variables. Associations of CVD risk factors and clinical data, and lipid profile with low ABI were assessed using logistic regression analysis. RESULTS The study participants had a mean age of 43.73 ± 8.74, majority were females (72.7%) with a mean duration on ART of 7.73 ± 3.52 years. Hypertension was present in 15.9%, diabetes 4%, family history of CVD 8.6% and metabolic syndrome 17.3% in the study group. The study participants recorded significantly lower mean values for ABI, HDL-C and significantly higher mean values of TG (P < 0.05) compared to the control group. The prevalence of low ABI (14.6%) was higher in the study group compared to the control group (2%). A significantly negative correlation between ABI and duration on ART (r = - 0.163, P = 0.041) and a positive correlation between viral load and TG were observed in the study group. TC (OR 1.784, P = 0.011), LDL-C (OR 1.824, P = 0.010) and CD4 cell count < 200 cells/mm3 (OR 2.635, P = 0.364) were associated with low ABI in the participants. CONCLUSION Viral suppression with combined antiretroviral therapy and long term treatment is associated with dyslipidaemia, with increased risk of PAD. Prevalence of PAD in virologically-suppressed individuals does not differ from the controls in the population studied.
Collapse
Affiliation(s)
- Chidozie Elochukwu Agu
- Prime Health Response Initiative (PHRI)-sub-recipients of Global Fund HIV Impact Project, Ilorin, Kwara State Nigeria
| | - Ikenna Kingsley Uchendu
- Department of Medical Laboratory Science, Faculty of Health Science and Technology, College of Medicine, University of Nigeria Enugu Campus, Enugu, Nigeria
| | - Augusta Chinyere Nsonwu
- Department of Medical Laboratory Science, Faculty of Allied Medical Science, University of Calabar, Calabar, Cross River State Nigeria
| | - Chukwugozie Nwachukwu Okwuosa
- Department of Medical Laboratory Science, Faculty of Health Science and Technology, College of Medicine, University of Nigeria Enugu Campus, Enugu, Nigeria
| | - Peter Uwadiegwu Achukwu
- Department of Medical Laboratory Science, Faculty of Health Science and Technology, College of Medicine, University of Nigeria Enugu Campus, Enugu, Nigeria
| |
Collapse
|
124
|
Kang I, Park M, Yang SJ, Lee M. Lipoprotein Lipase Inhibitor, Nordihydroguaiaretic Acid, Aggravates Metabolic Phenotypes and Alters HDL Particle Size in the Western Diet-Fed db/db Mice. Int J Mol Sci 2019; 20:ijms20123057. [PMID: 31234537 PMCID: PMC6627211 DOI: 10.3390/ijms20123057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 12/19/2022] Open
Abstract
Lipoprotein lipase (LPL) hydrolyzes triglycerides in lipoprotein to supply fatty acids, and its deficiency leads to hypertriglyceridemia, thereby inducing metabolic syndrome (MetSyn). Nordihydroguaiaretic acid (NDGA) has been recently reported to inhibit LPL secretion by endoplasmic reticulum (ER)-Golgi redistribution. However, the role of NDGA on dyslipidemia and MetSyn remains unclear. To address this question, leptin receptor knock out (KO)-db/db mice were randomly assigned to three different groups: A normal AIN76-A diet (CON), a Western diet (WD) and a Western diet with 0.1% NDGA and an LPL inhibitor, (WD+NDGA). All mice were fed for 12 weeks. The LPL inhibition by NDGA was confirmed by measuring the systemic LPL mass and adipose LPL gene expression. We investigated whether the LPL inhibition by NDGA alters the metabolic phenotypes. NDGA led to hyperglycemia, hypertriglyceridemia, and hypercholesterolemia. More strikingly, the supplementation of NDGA increased the percentage of high density lipoprotein (HDL)small (HDL3a+3b+3c) and decreased the percentage of HDLlarge (HDL2a+2b) compared to the WD group, which indicates that LPL inhibition modulates HDL subclasses. was NDGA increased adipose inflammation but had no impact on hepatic stress signals. Taken together, these findings demonstrated that LPL inhibition by NDGA aggravates metabolic parameters and alters HDL particle size.
Collapse
Affiliation(s)
- Inhae Kang
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Korea.
| | - Miyoung Park
- Research Institute of Obesity Sciences, Sungshin Women's University, Seoul 01133, Korea.
| | - Soo Jin Yang
- Department of Food and Nutrition, Seoul Women's University, Seoul 01797, Korea.
| | - Myoungsook Lee
- Research Institute of Obesity Sciences, Sungshin Women's University, Seoul 01133, Korea.
- Department of Food and Nutrition, Sungshin Women's University, Seoul 01133, Korea.
| |
Collapse
|
125
|
Pirillo A, Catapano AL, Norata GD. Biological Consequences of Dysfunctional HDL. Curr Med Chem 2019; 26:1644-1664. [PMID: 29848265 DOI: 10.2174/0929867325666180530110543] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/25/2017] [Accepted: 12/27/2017] [Indexed: 12/31/2022]
Abstract
Epidemiological studies have suggested an inverse correlation between high-density lipoprotein (HDL) cholesterol levels and the risk of cardiovascular disease. HDLs promote reverse cholesterol transport (RCT) and possess several putative atheroprotective functions, associated to the anti-inflammatory, anti-thrombotic and anti-oxidant properties as well as to the ability to support endothelial physiology. The assumption that increasing HDL-C levels would be beneficial on cardiovascular disease (CVD), however, has been questioned as, in most clinical trials, HDL-C-raising therapies did not result in improved cardiovascular outcomes. These findings, together with the observations from Mendelian randomization studies showing that polymorphisms mainly or solely associated with increased HDL-C levels did not decrease the risk of myocardial infarction, shift the focus from HDL-C levels toward HDL functional properties. Indeed, HDL from atherosclerotic patients not only exhibit impaired atheroprotective functions but also acquire pro-atherogenic properties and are referred to as "dysfunctional" HDL; this occurs even in the presence of normal or elevated HDL-C levels. Pharmacological approaches aimed at restoring HDL functions may therefore impact more significantly on CVD outcome than drugs used so far to increase HDL-C levels. The aim of this review is to discuss the pathological conditions leading to the formation of dysfunctional HDL and their role in atherosclerosis and beyond.
Collapse
Affiliation(s)
- Angela Pirillo
- Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy.,IRCCS Multimedica, Milan, Italy
| | - Alberico Luigi Catapano
- IRCCS Multimedica, Milan, Italy.,Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giuseppe Danilo Norata
- Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy.,Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.,School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia
| |
Collapse
|
126
|
Bashore AC, Liu M, Key CCC, Boudyguina E, Wang X, Carroll CM, Sawyer JK, Mullick AE, Lee RG, Macauley SL, Parks JS. Targeted Deletion of Hepatocyte Abca1 Increases Plasma HDL (High-Density Lipoprotein) Reverse Cholesterol Transport via the LDL (Low-Density Lipoprotein) Receptor. Arterioscler Thromb Vasc Biol 2019; 39:1747-1761. [PMID: 31167565 DOI: 10.1161/atvbaha.119.312382] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE The role of hepatocyte Abca1 (ATP binding cassette transporter A1) in trafficking hepatic free cholesterol (FC) into plasma versus bile for reverse cholesterol transport (RCT) is poorly understood. We hypothesized that hepatocyte Abca1 recycles plasma HDL-C (high-density lipoprotein cholesterol) taken up by the liver back into plasma, maintaining the plasma HDL-C pool, and decreasing HDL-mediated RCT into feces. Approach and Results: Chow-fed hepatocyte-specific Abca1 knockout (HSKO) and control mice were injected with human HDL radiolabeled with 125I-tyramine cellobiose (125I-TC; protein) and 3H-cholesteryl oleate (3H-CO). 125I-TC and 3H-CO plasma decay, plasma HDL 3H-CO selective clearance (ie, 3H-125I fractional catabolic rate), liver radiolabel uptake, and fecal 3H-sterol were significantly greater in HSKO versus control mice, supporting increased plasma HDL RCT. Twenty-four hours after 3H-CO-HDL injection, HSKO mice had reduced total hepatic 3H-FC (ie, 3H-CO hydrolyzed to 3H-FC in liver) resecretion into plasma, demonstrating Abca1 recycled HDL-derived hepatic 3H-FC back into plasma. Despite similar liver LDLr (low-density lipoprotein receptor) expression between genotypes, HSKO mice treated with LDLr-targeting versus control antisense oligonucleotide had slower plasma 3H-CO-HDL decay, reduced selective 3H-CO clearance, and decreased fecal 3H-sterol excretion that was indistinguishable from control mice. Increased RCT in HSKO mice was selective for 3H-CO-HDL, since macrophage RCT was similar between genotypes. CONCLUSIONS Hepatocyte Abca1 deletion unmasks a novel and selective FC trafficking pathway that requires LDLr expression, accelerating plasma HDL-selective CE uptake by the liver and promoting HDL RCT into feces, consequently reducing HDL-derived hepatic FC recycling into plasma.
Collapse
Affiliation(s)
- Alexander C Bashore
- From the Department of Internal Medicine, Section of Molecular Medicine (A.C.B., M.L., C-C.C.K., E.B., X.W., J.K.S., J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC
| | - Mingxia Liu
- From the Department of Internal Medicine, Section of Molecular Medicine (A.C.B., M.L., C-C.C.K., E.B., X.W., J.K.S., J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC
| | - Chia-Chi C Key
- From the Department of Internal Medicine, Section of Molecular Medicine (A.C.B., M.L., C-C.C.K., E.B., X.W., J.K.S., J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC
| | - Elena Boudyguina
- From the Department of Internal Medicine, Section of Molecular Medicine (A.C.B., M.L., C-C.C.K., E.B., X.W., J.K.S., J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC
| | - Xianfeng Wang
- From the Department of Internal Medicine, Section of Molecular Medicine (A.C.B., M.L., C-C.C.K., E.B., X.W., J.K.S., J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC
| | - Caitlin M Carroll
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine (C.M.C., S.L.M.), Wake Forest School of Medicine, Winston-Salem, NC
| | - Janet K Sawyer
- From the Department of Internal Medicine, Section of Molecular Medicine (A.C.B., M.L., C-C.C.K., E.B., X.W., J.K.S., J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC
| | - Adam E Mullick
- Cardiovascular, Renal and Metabolic Group, Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M., R.G.L.)
| | - Richard G Lee
- Cardiovascular, Renal and Metabolic Group, Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (A.E.M., R.G.L.)
| | - Shannon L Macauley
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine (C.M.C., S.L.M.), Wake Forest School of Medicine, Winston-Salem, NC
| | - John S Parks
- From the Department of Internal Medicine, Section of Molecular Medicine (A.C.B., M.L., C-C.C.K., E.B., X.W., J.K.S., J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC
| |
Collapse
|
127
|
Bardagjy AS, Steinberg FM. Relationship Between HDL Functional Characteristics and Cardiovascular Health and Potential Impact of Dietary Patterns: A Narrative Review. Nutrients 2019; 11:E1231. [PMID: 31151202 PMCID: PMC6627343 DOI: 10.3390/nu11061231] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/25/2019] [Accepted: 05/27/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is a leading cause of death around the world. Overall diet quality and dietary behaviors are core contributors to metabolic health. While therapeutic targets have traditionally focused on levels of lipoprotein cholesterol when evaluating cardiovascular risk, current perspectives on high-density lipoprotein (HDL) have shifted to evaluating the functionality of this lipoprotein particle. Effects of diet on cardiovascular health are mediated through multiple pathways, but the impact on HDL composition and function deserves greater attention. Potential areas of investigation involve changes in particle characteristics, distribution, microRNA cargo, and other functional changes such as improvements to cholesterol efflux capacity. Various dietary patterns like the Mediterranean diet and Dietary Approaches to Stop Hypertension (DASH) diet have beneficial effects on cardiovascular health and may prevent cardiovascular events. These healthful dietary patterns tend to be rich in plant-based foods, with cardiovascular benefits likely resulting from synergistic effects of the individual dietary components. The purpose of this review is to summarize current perspectives on selected functions of HDL particles and how various dietary patterns affect cardiovascular health biomarkers, with a focus on HDL functionality.
Collapse
Affiliation(s)
- Allison S Bardagjy
- Department of Nutrition and Graduate Group in Nutritional Biology, University of California, Davis, CA 95616, USA.
| | - Francene M Steinberg
- Department of Nutrition and Graduate Group in Nutritional Biology, University of California, Davis, CA 95616, USA.
| |
Collapse
|
128
|
Cui Y, Liu J, Huang C, Zhao B. Moxibustion at CV4 alleviates atherosclerotic lesions through activation of the LXRα/ABCA1 pathway in apolipoprotein-E-deficient mice. Acupunct Med 2019; 37:237-243. [PMID: 31140825 PMCID: PMC7433780 DOI: 10.1136/acupmed-2016-011317] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Objectives: To investigate the anti-atherogenic effect of moxibustion and whether it is mediated through the reverse cholesterol transport process. Methods: 8-week-old male apolipoprotein E deficient (ApoE−/− knockout) mice were randomly divided into two groups (n=10 per group): atherosclerosis (AS) and AS plus moxibustion (AS+M). C57BL/6J mice of the same background (n=10) were selected as controls. Mice in the AS+M group received indirect moxibustion with an ignited moxa stick held over CV4. Mice of the AS and control groups were restrained in the same holder with an unlit moxa stick held over CV4. All treatments were performed for 20 min per day, 6 days per week for 12 weeks. After the treatment, the mice were euthanased and their serum lipids were measured. The aortic roots and thoracic aortas were collected for haematoxylin and eosin and red oil O staining, respectively, to analyse the atherosclerotic lesions. Expression of adenosine triphosphate binding cassette (ABCA)A1/G1 and liver X receptor α (LXRα) in the thoracic aorta were examined with Western blotting. Results: The moxibustion-treated (AS+M) mice showed a significantly lower plaque area percentage in the aortic root and thoracic aorta, and higher expression of LXRα and ABCA1 in the thoracic aorta compared with the AS mice. No significant differences were found in average lipid area percentage in the thoracic aorta, or ABCG1 expression in the thoracic aorta, between mice in the AS+M and AS groups. Conclusion: Moxibustion treatment at CV4 suppressed the progression of atherosclerotic lesions in ApoE−/− mice. The anti-atherogenic effect of moxibustion may be achieved by: (1) regulation of lipid metabolism, and thus prevention of lipid accumulation; and (2) upregulation of LXRα- and ABCA1-mediated cholesterol efflux in the lesion area.
Collapse
Affiliation(s)
- Yingxue Cui
- 1 Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Juntian Liu
- 2 Acupuncture and Moxibustion Department, Huguosi Hospital of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chang Huang
- 3 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Baixiao Zhao
- 3 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
129
|
Ossoli A, Pavanello C, Giorgio E, Calabresi L, Gomaraschi M. Dysfunctional HDL as a Therapeutic Target for Atherosclerosis Prevention. Curr Med Chem 2019; 26:1610-1630. [DOI: 10.2174/0929867325666180316115726] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/24/2017] [Accepted: 12/26/2017] [Indexed: 12/12/2022]
Abstract
Hypercholesterolemia is one of the main risk factors for the development of atherosclerosis. Among the various lipoprotein classes, however, high density lipoproteins (HDL) are inversely associated with the incidence of atherosclerosis, since they are able to exert a series of atheroprotective functions. The central role of HDL within the reverse cholesterol transport, their antioxidant and anti-inflammatory properties and their ability to preserve endothelial homeostasis are likely responsible for HDL-mediated atheroprotection. However, drugs that effectively raise HDL-C failed to result in a decreased incidence of cardiovascular event, suggesting that plasma levels of HDL-C and HDL function are not always related. Several evidences are showing that different pathologic conditions, especially those associated with an inflammatory response, can cause dramatic alterations of HDL protein and lipid cargo resulting in HDL dysfunction. Established and investigational drugs designed to affect lipid metabolism and to increase HDL-C are only partly effective in correcting HDL dysfunction.
Collapse
Affiliation(s)
- Alice Ossoli
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Chiara Pavanello
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Eleonora Giorgio
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Laura Calabresi
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Monica Gomaraschi
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
130
|
Maric S, Lind TK, Raida MR, Bengtsson E, Fredrikson GN, Rogers S, Moulin M, Haertlein M, Forsyth VT, Wenk MR, Pomorski TG, Arnebrant T, Lund R, Cárdenas M. Time-resolved small-angle neutron scattering as a probe for the dynamics of lipid exchange between human lipoproteins and naturally derived membranes. Sci Rep 2019; 9:7591. [PMID: 31110185 PMCID: PMC6527577 DOI: 10.1038/s41598-019-43713-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/27/2019] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis is the main killer in the western world. Today’s clinical markers include the total level of cholesterol and high-/low-density lipoproteins, which often fails to accurately predict the disease. The relationship between the lipid exchange capacity and lipoprotein structure should explain the extent by which they release or accept lipid cargo and should relate to the risk for developing atherosclerosis. Here, small-angle neutron scattering and tailored deuteration have been used to follow the molecular lipid exchange between human lipoprotein particles and cellular membrane mimics made of natural, “neutron invisible” phosphatidylcholines. We show that lipid exchange occurs via two different processes that include lipid transfer via collision and upon direct particle tethering to the membrane, and that high-density lipoprotein excels at exchanging the human-like unsaturated phosphatidylcholine. By mapping the specific lipid content and level of glycation/oxidation, the mode of action of specific lipoproteins can now be deciphered. This information can prove important for the development of improved diagnostic tools and in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Selma Maric
- Dept. of Biomedical Science, Malmö University, Per Albin Hanssons väg 35, 205 02, Malmö, Sweden.
| | - Tania Kjellerup Lind
- Dept. of Biomedical Science, Malmö University, Per Albin Hanssons väg 35, 205 02, Malmö, Sweden
| | - Manfred Roman Raida
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Eva Bengtsson
- Dept. of Clinical Sciences, Lund University, Jan Waldenströms gata 35, CRC, Box 50332, 212 13, Malmö, Sweden
| | - Gunilla Nordin Fredrikson
- Dept. of Clinical Sciences, Lund University, Jan Waldenströms gata 35, CRC, Box 50332, 212 13, Malmö, Sweden
| | - Sarah Rogers
- ISIS Science and Technology Facilities Council, Harwell Science and Innovation Campus, Chilton, Didcot, Oxfordshire, OX11 0QX, United Kingdom
| | - Martine Moulin
- Life Science Group, Institut Laue Langevin, 6, rue Jules Horowitz, BP 156, F-38042, Grenoble, Cedex 9, France
| | - Michael Haertlein
- Life Science Group, Institut Laue Langevin, 6, rue Jules Horowitz, BP 156, F-38042, Grenoble, Cedex 9, France
| | - V Trevor Forsyth
- Life Science Group, Institut Laue Langevin, 6, rue Jules Horowitz, BP 156, F-38042, Grenoble, Cedex 9, France.,Faculty of Natural Science and Institute for Science and Technology in Medicine, Keele University, Staffordshire, ST5 5BG, United Kingdom
| | - Markus R Wenk
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Thomas Günther Pomorski
- Dept. of Plant and Environmental Sciences, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg C, Denmark.,Dept. of Molecular Biochemistry, Ruhr University Bochum, Faculty of Chemistry and Biochemistry, 44780, Bochum, Germany
| | - Thomas Arnebrant
- Dept. of Biomedical Science, Malmö University, Per Albin Hanssons väg 35, 205 02, Malmö, Sweden
| | - Reidar Lund
- Dept. of Chemistry, University of Oslo, Blindern, 0315, Oslo, Norway
| | - Marité Cárdenas
- Dept. of Biomedical Science, Malmö University, Per Albin Hanssons väg 35, 205 02, Malmö, Sweden.
| |
Collapse
|
131
|
Matsuo N, Nagao K, Suito T, Juni N, Kato U, Hara Y, Umeda M. Different mechanisms for selective transport of fatty acids using a single class of lipoprotein in Drosophila. J Lipid Res 2019; 60:1199-1211. [PMID: 31085629 DOI: 10.1194/jlr.m090779] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 05/10/2019] [Indexed: 12/16/2022] Open
Abstract
In mammals, lipids are selectively transported to specific sites using multiple classes of lipoproteins. However, in Drosophila, a single class of lipoproteins, lipophorin, carries more than 95% of the lipids in the hemolymph. Although a unique ability of the insect lipoprotein system for cargo transport has been demonstrated, it remains unclear how this single class of lipoproteins selectively transports lipids. In this study, we carried out a comparative analysis of the fatty-acid composition among lipophorin, the CNS, and CNS-derived cell lines and investigated the transport mechanism of fatty acids, particularly focusing on the transport of PUFAs in Drosophila We showed that PUFAs are selectively incorporated into the acyl chains of lipophorin phospholipids and effectively transported to CNS through lipophorin receptor-mediated endocytosis of lipophorin. In addition, we demonstrated that C14 fatty acids are selectively incorporated into the diacylglycerols (DAGs) of lipophorin and that C14 fatty-acid-containing DAGs are spontaneously transferred from lipophorin to the phospholipid bilayer. These results suggest that PUFA-containing phospholipids and C14 fatty-acid-containing DAGs in lipophorin could be transferred to different sites by different mechanisms to selectively transport fatty acids using a single class of lipoproteins.
Collapse
Affiliation(s)
- Naoya Matsuo
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Kohjiro Nagao
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Takuto Suito
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Naoto Juni
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Utako Kato
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Yuji Hara
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan.,AMED-PRIME Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| | - Masato Umeda
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| |
Collapse
|
132
|
Wu C, Wang Y, Gong P, Wang L, Liu C, Chen C, Jiang X, Dong X, Cheng B, Li H. Promoter Methylation Regulates ApoA-I Gene Transcription in Chicken Abdominal Adipose Tissue. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:4535-4544. [PMID: 30932484 DOI: 10.1021/acs.jafc.9b00007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
As a central constituent of HDL (high-density lipoprotein), apolipoprotein A-I (ApoA-I) has a vital function in lipid metabolism. Our previous studies confirmed that ApoA-I was differentially expressed in the adipose tissue of the abdomen of lean and fat broilers. The aim of the current work was to evaluate whether the transcription of ApoA-I in chicken abdominal adipose tissue was regulated by DNA methylation. The methylation status of ApoA-I promoter CpG island (PCGI) and promoter non-CpG island (PNCGI) as well as the ApoA-I expression level in adipose tissue of lean and fat broilers were determined using Sequenom MassARRAY and real-time PCR. The correlation analysis results showed that the methylation level of PCGI and the ApoA-I mRNA expression level were negatively correlated. Bisulfite sequencing PCR was used to assess the methylation level of ApoA-I promoter in the ICP1 cells treated with 5-aza-2'-deoxycytidine (5-Aza-CdR: an inhibitor of DNA methyltransferase). The result showed that 5-Aza-CdR caused a reduction in the methylation level of the ApoA-I promoter, thereby causing an increase in expression of the ApoA-I mRNA. Meanwhile, luciferase reporter assays indicated that in vitro methylation of the ApoA-I promoter containing CpG island with CpG methyltransferase led to transcriptional repression. Furthermore, the noticeable activation of NRF1 on ApoA-I transcription was largely enhanced by the demethylation of the ApoA-I PCGI region. These observations indicated that the differential expression of ApoA-I gene in the adipose tissue of broilers could be mediated by transcription regulation, at least in part by DNA methylation in its PCGI region.
Collapse
Affiliation(s)
- Chunyan Wu
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction of Education Department of Heilongjiang Province, College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Yuxiang Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction of Education Department of Heilongjiang Province, College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Pengfei Gong
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction of Education Department of Heilongjiang Province, College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Lijian Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction of Education Department of Heilongjiang Province, College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Chang Liu
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction of Education Department of Heilongjiang Province, College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Chong Chen
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction of Education Department of Heilongjiang Province, College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Xiuying Jiang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction of Education Department of Heilongjiang Province, College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Xiangyu Dong
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction of Education Department of Heilongjiang Province, College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Bohan Cheng
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction of Education Department of Heilongjiang Province, College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Hui Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Genetics, Breeding and Reproduction of Education Department of Heilongjiang Province, College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| |
Collapse
|
133
|
Dash S, Leiter LA. Residual cardiovascular risk among people with diabetes. Diabetes Obes Metab 2019; 21 Suppl 1:28-38. [PMID: 31002458 DOI: 10.1111/dom.13646] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/16/2019] [Accepted: 01/28/2019] [Indexed: 01/05/2023]
Abstract
Type 2 diabetes (T2D) is a growing health concern across both developed and developing countries. Cardiovascular disease (CVD) remains the major cause of increased mortality in this patient population. In recent years, effective low density lipoprotein lowering treatments and other risk reduction strategies have substantially reduced the risk of atherosclerotic CVD, yet patients with T2D continue to remain at increased risk for atherosclerotic CVD. Here, we will briefly review various proposed underlying mechanisms for this residual risk with a more in-depth focus on the potential role of triglyceride-rich lipoproteins in residual risk and potential avenues to target this pharmacologically.
Collapse
Affiliation(s)
- Satya Dash
- Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | - Lawrence A Leiter
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
134
|
Fazel F, Naghibzadeh A, Mohammad Ramezanpour R, Bagheri R, Hamidi A, Rashidlamir A. Effect of Aerobic Exercise with and without Green Coffee Supplementation on Serum Apolipoprotein B and Atherogenic Indices of Overweight Men. MEDICAL LABORATORY JOURNAL 2019. [DOI: 10.29252/mlj.13.3.20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
135
|
Holzer M, Marsche G. Comment on Mathew et al. Therapeutic Lifestyle Changes Improve HDL Function by Inhibiting Myeloperoxidase-Mediated Oxidation in Patients With Metabolic Syndrome. Diabetes Care 2018;41:2431-2437. Diabetes Care 2019; 42:e25. [PMID: 30665968 DOI: 10.2337/dc18-2026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Michael Holzer
- Division of Pharmacology, Otto Loewi Research Centre, Medical University of Graz, Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Centre, Medical University of Graz, Graz, Austria.,BioTechMed-Graz, Graz, Austria
| |
Collapse
|
136
|
Rauschert S, Gázquez A, Uhl O, Kirchberg FF, Demmelmair H, Ruíz-Palacios M, Prieto-Sánchez MT, Blanco-Carnero JE, Nieto A, Larqué E, Koletzko B. Phospholipids in lipoproteins: compositional differences across VLDL, LDL, and HDL in pregnant women. Lipids Health Dis 2019; 18:20. [PMID: 30670033 PMCID: PMC6343318 DOI: 10.1186/s12944-019-0957-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 01/01/2019] [Indexed: 12/11/2022] Open
Abstract
Objective The aim of this study was to analyse the differences in the phospholipid composition of very low density (VLDL), low density (LDL) and high density lipoprotein (HDL) monolayers in pregnant lean and obese women. Methods LDL, HDL, and VLDL were isolated from plasma samples of 10 lean and 10 obese pregnant women, and their species composition of phosphatidylcholines (PC) and sphingomyelins (SM) was analysed by liquid-chromatography tandem mass-spectrometry. Wilcoxon-Mann-Whitney U test and principal component analysis (PCA) were used to investigate if metabolite profiles differed between the lean/obese group and between lipoprotein species. Results No significant differences have been found in the metabolite levels between obese and non-obese pregnant women. The PCA components 1 and 2 separated between LDL, HDL, and VLDL but not between normal weight and obese women. Twelve SM and one PCae were more abundant in LDL than in VLDL. In contrast, four acyl-alkyl-PC and two diacyl-PC were significantly higher in HDL compared to LDL. VLDL and HDL differed in three SM, seven acyl-alkyl-PC and one diacyl-PC (higher values in HDL) and 13 SM (higher in VLDL). We also found associations of some phospholipid species with HDL and LDL cholesterol. Conclusion In pregnant women phospholipid composition differs significantly in HDL, LDL and VLDL, similar to previous findings in men and non-pregnant women. Obese and lean pregnant women showed no significant differences in their lipoprotein associated metabolite profile. Electronic supplementary material The online version of this article (10.1186/s12944-019-0957-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sebastian Rauschert
- LMU - Ludwig-Maximilians-Universität Munich, Div. Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337, Munich, Germany
| | - Antonio Gázquez
- LMU - Ludwig-Maximilians-Universität Munich, Div. Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337, Munich, Germany.,Department of Physiology, Faculty of Biology, University of Murcia, Murcia, Spain
| | - Olaf Uhl
- LMU - Ludwig-Maximilians-Universität Munich, Div. Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337, Munich, Germany
| | - Franca F Kirchberg
- LMU - Ludwig-Maximilians-Universität Munich, Div. Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337, Munich, Germany
| | - Hans Demmelmair
- LMU - Ludwig-Maximilians-Universität Munich, Div. Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337, Munich, Germany
| | - María Ruíz-Palacios
- Department of Physiology, Faculty of Biology, University of Murcia, Murcia, Spain
| | - María T Prieto-Sánchez
- Obstetrics and Gynecology Service, Virgen de la Arrixaca Clinical Hospital, University of Murcia, Murcia, Murcia, Spain
| | - José E Blanco-Carnero
- Obstetrics and Gynecology Service, Virgen de la Arrixaca Clinical Hospital, University of Murcia, Murcia, Murcia, Spain
| | - Anibal Nieto
- Obstetrics and Gynecology Service, Virgen de la Arrixaca Clinical Hospital, University of Murcia, Murcia, Murcia, Spain
| | - Elvira Larqué
- Department of Physiology, Faculty of Biology, University of Murcia, Murcia, Spain
| | - Berthold Koletzko
- LMU - Ludwig-Maximilians-Universität Munich, Div. Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337, Munich, Germany.
| |
Collapse
|
137
|
Yang S, Yin RX, Miao L, Zhang QH, Zhou YG, Wu J. Association between the LIPG polymorphisms and serum lipid levels in the Maonan and Han populations. J Gene Med 2019; 21:e3071. [PMID: 30657227 PMCID: PMC6590183 DOI: 10.1002/jgm.3071] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/21/2018] [Accepted: 12/24/2018] [Indexed: 01/19/2023] Open
Abstract
Introduction The Maonan population is a relatively isolated minority in China. Little is known about endothelial lipase gene (LIPG) single nucleotide polymorphisms (SNPs) and serum lipid levels in the Chinese populations. The present study aimed to detect the association of several LIPG SNPs and environmental factors with serum lipid levels in the Chinese Maonan and Han populations. Methods In total, 773 subjects of Maonan ethnicity and 710 participants of Han ethnicity were randomly selected from our previous stratified randomized samples. Genotypes of the LIPG rs2156552, rs4939883 and rs7241918 SNPs were determined by polymerase chain reaction‐restriction fragment length polymorphism, and then confirmed by direct sequencing. Results The allelic (rs2156552, rs4939883 and rs7241918) and genotypic (rs2156552 and rs4939883) frequencies were different between the two ethnic groups (p < 0.05–0.01). The minor allele carriers had lower apolipoprotein (Apo)A1/ApoB ratio (rs2156552 and rs7241918), high‐density lipoprotein cholesterol (HDL‐C) and apolipoprotein (Apo)A1 (rs2156552) levels and higher ApoB levels (rs4939883) in the Han population, and lower HDL‐C (rs2156552, rs4939883 and rs7241918) levels in the Maonan minority than the minor allele non‐carriers (p < 0.0167 after Bonferroni correction). Subgroup analyses according to sex showed that the minor allele carriers had a lower ApoA1/ApoB ratio (rs2156552 and rs7241918) and higher ApoB levels (rs7241918) in Han males, and lower ApoA1 and HDL‐C levels in Maonan females than the minor allele non‐carriers (p < 0.0167–0.001). Conclusions The present study demonstrates the association between the LIPG polymorphsims and serum lipid levels in the two ethnic groups. These associations might have an ethnic‐ and or/sex‐specificity.
Collapse
Affiliation(s)
- Shuo Yang
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Rui-Xing Yin
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Liu Miao
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Qing-Hui Zhang
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Yong-Gang Zhou
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Jie Wu
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
138
|
Maylor BD, Zakrzewski-Fruer JK, Orton CJ, Bailey DP. Beneficial postprandial lipaemic effects of interrupting sedentary time with high-intensity physical activity versus a continuous moderate-intensity physical activity bout: A randomised crossover trial. J Sci Med Sport 2018; 21:1250-1255. [DOI: 10.1016/j.jsams.2018.05.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 05/14/2018] [Accepted: 05/24/2018] [Indexed: 12/20/2022]
|
139
|
Knowles EEM, Curran JE, Meikle PJ, Huynh K, Mathias SR, Göring HHH, VandeBerg JL, Mahaney MC, Jalbrzikowski M, Mosior MK, Michael LF, Olvera RL, Duggirala R, Almasy L, Glahn DC, Blangero J. Disentangling the genetic overlap between cholesterol and suicide risk. Neuropsychopharmacology 2018; 43:2556-2563. [PMID: 30082891 PMCID: PMC6224547 DOI: 10.1038/s41386-018-0162-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 01/03/2023]
Abstract
Suicide is major public health concern; one million individuals worldwide die by suicide each year of which there are many more attempts. Thus, it is imperative that robust and reliable indicators, or biomarkers, of suicide risk be identified so that individuals at risk can be identified and provided appropriate interventions as quickly as possible. Previous work has revealed a relationship between low levels of circulating cholesterol and suicide risk, implicating cholesterol level as one such potential biomarker, but the factors underlying this relationship remain unknown. In the present study, we applied a combination of bivariate polygenic and coefficient-of-relatedness analysis, followed by mediation analysis, in a large sample of Mexican-American individuals from extended pedigrees [N = 1897; 96 pedigrees (average size = 19.17 individuals, range = 2-189) 60% female; mean age = 42.58 years, range = 18-97 years, sd = 15.75 years] with no exclusion criteria for any given psychiatric disorder. We observed that total esterified cholesterol measured at the time of psychiatric assessment shared a significant genetic overlap with risk for suicide attempt (ρg = -0.64, p = 1.24 × 10-04). We also found that total unesterified cholesterol measured around 20 years prior to assessment varied as a function of genetic proximity to an affected individual (h2 = 0.21, se = 0.10, p = 8.73 × 10-04; βsuicide = -0.70, se = 0.25, p = 8.90 × 10-03). Finally, we found that the relationship between total unesterified cholesterol and suicide risk was significantly mediated by ABCA-1-specific cholesterol efflux capacity (βsuicide-efflux = -0.45, p = 0.039; βefflux-cholexterol = -0.34, p < 0.0001; βindirect = -0.15, p = 0.044). These findings suggest that the relatively well-delineated process of cholesterol metabolism and associated molecular pathways will be informative for understanding the neurobiological underpinnings of risk for suicide attempt.
Collapse
Affiliation(s)
- Emma E. M. Knowles
- 0000000419368710grid.47100.32Department of Psychiatry, Yale University School of Medicine, New Haven, CT USA
| | - Joanne E. Curran
- 0000 0004 5374 269Xgrid.449717.8South Texas Diabetes and Obesity Institute and Department of Human Genetics, University of Texas of the Rio Grande Valley School of Medicine, Brownsville, TX USA
| | - Peter J. Meikle
- 0000 0000 9760 5620grid.1051.5Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Kevin Huynh
- 0000 0000 9760 5620grid.1051.5Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Samuel R. Mathias
- 0000000419368710grid.47100.32Department of Psychiatry, Yale University School of Medicine, New Haven, CT USA
| | - Harald H. H. Göring
- 0000 0004 5374 269Xgrid.449717.8South Texas Diabetes and Obesity Institute and Department of Human Genetics, University of Texas of the Rio Grande Valley School of Medicine, Brownsville, TX USA
| | - John L. VandeBerg
- 0000 0004 5374 269Xgrid.449717.8South Texas Diabetes and Obesity Institute and Department of Human Genetics, University of Texas of the Rio Grande Valley School of Medicine, Brownsville, TX USA
| | - Michael C. Mahaney
- 0000 0004 5374 269Xgrid.449717.8South Texas Diabetes and Obesity Institute and Department of Human Genetics, University of Texas of the Rio Grande Valley School of Medicine, Brownsville, TX USA
| | - Maria Jalbrzikowski
- 0000 0004 1936 9000grid.21925.3dDepartment of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA
| | - Marian K. Mosior
- 0000 0000 2220 2544grid.417540.3Diabetes and Complications Therapeutic Area, Eli Lilly and Company, Indianapolis, IN USA
| | - Laura F. Michael
- 0000 0000 2220 2544grid.417540.3Diabetes and Complications Therapeutic Area, Eli Lilly and Company, Indianapolis, IN USA
| | - Rene L. Olvera
- 0000 0001 0629 5880grid.267309.9Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX USA
| | - Ravi Duggirala
- 0000 0004 5374 269Xgrid.449717.8South Texas Diabetes and Obesity Institute and Department of Human Genetics, University of Texas of the Rio Grande Valley School of Medicine, Brownsville, TX USA
| | - Laura Almasy
- 0000 0004 1936 8972grid.25879.31Department of Genetics, University of Pennsylvania, Philadelphia, PA USA ,0000 0001 0680 8770grid.239552.aDepartment of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA USA
| | - David C. Glahn
- 0000000419368710grid.47100.32Department of Psychiatry, Yale University School of Medicine, New Haven, CT USA ,0000 0001 0626 2712grid.277313.3Olin Neuropsychiatric Research Center, Institute of Living, Hartford Hospital, Hartford, CT USA
| | - John Blangero
- 0000 0004 5374 269Xgrid.449717.8South Texas Diabetes and Obesity Institute and Department of Human Genetics, University of Texas of the Rio Grande Valley School of Medicine, Brownsville, TX USA
| |
Collapse
|
140
|
Ray A, Ghosh A, Chakraborty R, Upadhyay SK, Maiti S, Sengupta S, Thukral L. Specific Cholesterol Binding Drives Drastic Structural Alterations in Apolipoprotein A1. J Phys Chem Lett 2018; 9:6060-6065. [PMID: 30256643 DOI: 10.1021/acs.jpclett.8b02042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Proteins typically adopt a multitude of flexible and rapidly interconverting conformers, many of which are governed by specific protein interaction domains. Whereas disc-shaped oligomeric HDL and its major protein component ApoA1 have been the focus of several investigations, the structural properties of monomeric ApoA1 remain poorly understood. Using tens of independent molecular simulations (>50 μs), we reveal that ApoA1 adopts a compact conformation. Upon the addition of a physiological concentration of cholesterol to ApoA1, the monomeric protein spontaneously formed a circular conformation. Remarkably, these drastic structural perturbations are driven by a specific cholesterol binding site at the C-terminal and a novel cholesterol binding site at the N-terminal. We propose a mechanism whereby ApoA1 opens in a stagewise manner and mutating the N-terminal binding site destroys the open "belt-shaped" topology. Complementary experiments confirm that the structural changes are induced by specific association of cholesterol with ApoA1, not by the nonspecific hydrophobic effect.
Collapse
Affiliation(s)
- Arjun Ray
- CSIR-Institute of Genomics and Integrative Biology , South Campus, Mathura Road , New Delhi 110 025 , India
- Academy of Scientific and Innovative Research (AcSIR) , CSIR- Institute of Genomics and Integrative Biology , Mathura Road Campus , New Delhi 110025 , India
| | - Asmita Ghosh
- CSIR-Institute of Genomics and Integrative Biology , South Campus, Mathura Road , New Delhi 110 025 , India
- Academy of Scientific and Innovative Research (AcSIR) , CSIR- Institute of Genomics and Integrative Biology , Mathura Road Campus , New Delhi 110025 , India
| | - Rahul Chakraborty
- CSIR-Institute of Genomics and Integrative Biology , South Campus, Mathura Road , New Delhi 110 025 , India
- Academy of Scientific and Innovative Research (AcSIR) , CSIR- Institute of Genomics and Integrative Biology , Mathura Road Campus , New Delhi 110025 , India
| | - Santosh Kumar Upadhyay
- CSIR-Institute of Genomics and Integrative Biology , South Campus, Mathura Road , New Delhi 110 025 , India
| | - Souvik Maiti
- CSIR-Institute of Genomics and Integrative Biology , South Campus, Mathura Road , New Delhi 110 025 , India
- Academy of Scientific and Innovative Research (AcSIR) , CSIR- Institute of Genomics and Integrative Biology , Mathura Road Campus , New Delhi 110025 , India
| | - Shantanu Sengupta
- CSIR-Institute of Genomics and Integrative Biology , South Campus, Mathura Road , New Delhi 110 025 , India
- Academy of Scientific and Innovative Research (AcSIR) , CSIR- Institute of Genomics and Integrative Biology , Mathura Road Campus , New Delhi 110025 , India
| | - Lipi Thukral
- CSIR-Institute of Genomics and Integrative Biology , South Campus, Mathura Road , New Delhi 110 025 , India
- Academy of Scientific and Innovative Research (AcSIR) , CSIR- Institute of Genomics and Integrative Biology , Mathura Road Campus , New Delhi 110025 , India
- Interdisciplinary Center for Scientific Computing , University of Heidelberg , Im Neuenheimer Feld 205 , 69120 Heidelberg , Germany
| |
Collapse
|
141
|
Mao MG, Chen Y, Liu RT, Lü HQ, Gu J, Jiang ZQ, Jiang JL. Transcriptome from Pacific cod liver reveals types of apolipoproteins and expression analysis of AFP-IV, structural analogue with mammalian ApoA-I. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2018; 28:204-212. [PMID: 30366214 DOI: 10.1016/j.cbd.2018.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 10/04/2018] [Indexed: 12/15/2022]
Abstract
Apolipoproteins (Apos), transporting the lipids through the lymphatic and circulatory systems, are associated with kinds of diseases. Additionally, type IV antifreeze protein (AFP-IV) was related evolutionarily with apolipoproteins. However, the information of Apos in fish was limited. In this study, ApoA-I, ApoA-I-2, ApoA-IV, Apo E, ApoB-100-like and AFP-IV were sequenced from Pacific cod (Gadus macrocephalus) liver transcriptome using Illumina HiSeq 2000, and their 3-D models were constructed based on the most confidence templates ever reported in mammals. Interestingly, the model of G. macrocephalus AFP-IV, named GmAFPIV, is quite similar to the structure of ApoA-I. GmAFPIV includes 689 bases with a complete open reading frame encoding 125 amino acids. Sequence alignment of GmAFPIV showed 30% to 50% similarity with that of other species except Gadus sp. Expression levels of GmAFPIV were found in a decreasing manner in liver, intestine, gill, brain and gonad. Heterologously expression of the GmAFPIV protein was expressed in Escherichia coli and purified to immunize New Zealand rabbits. The survivors of E. coli in 60 μg/mL of GmAFPIV are more than that in the 30 μg/mL group after stored in -20 °C and -80 °C, indicating high concentration of GmAFPIV could protect E. coli avoiding the damage from ice crystal. The subcellular localization of GmAFPIV showed that the green fluorescence was mainly observed in the cytoplasm, indicating GmAFPIV play roles in the cytoplasm. It was concluded that GmAFPIV may function not only as an antifreeze protein but also as an apolipoprotein transporting lipids in fish.
Collapse
Affiliation(s)
- Ming-Guang Mao
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Key Laboratory of Fish Applied Biology and Aquaculture in North China, Liaoning Province, College of Fisheries and Life Sciences, Dalian Ocean University, Dalian 116023, China; State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Yu Chen
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Key Laboratory of Fish Applied Biology and Aquaculture in North China, Liaoning Province, College of Fisheries and Life Sciences, Dalian Ocean University, Dalian 116023, China
| | - Rui-Ting Liu
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Key Laboratory of Fish Applied Biology and Aquaculture in North China, Liaoning Province, College of Fisheries and Life Sciences, Dalian Ocean University, Dalian 116023, China
| | - Hui-Qian Lü
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Key Laboratory of Fish Applied Biology and Aquaculture in North China, Liaoning Province, College of Fisheries and Life Sciences, Dalian Ocean University, Dalian 116023, China
| | - Jie Gu
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Key Laboratory of Fish Applied Biology and Aquaculture in North China, Liaoning Province, College of Fisheries and Life Sciences, Dalian Ocean University, Dalian 116023, China
| | - Zhi-Qiang Jiang
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Key Laboratory of Fish Applied Biology and Aquaculture in North China, Liaoning Province, College of Fisheries and Life Sciences, Dalian Ocean University, Dalian 116023, China
| | - Jie-Lan Jiang
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Key Laboratory of Fish Applied Biology and Aquaculture in North China, Liaoning Province, College of Fisheries and Life Sciences, Dalian Ocean University, Dalian 116023, China.
| |
Collapse
|
142
|
Stamatikos A, Dronadula N, Ng P, Palmer D, Knight E, Wacker BK, Tang C, Kim F, Dichek DA. ABCA1 Overexpression in Endothelial Cells In Vitro Enhances ApoAI-Mediated Cholesterol Efflux and Decreases Inflammation. Hum Gene Ther 2018; 30:236-248. [PMID: 30079772 DOI: 10.1089/hum.2018.120] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis, a disease of blood vessels, is driven by cholesterol accumulation and inflammation. Gene therapy that removes cholesterol from blood vessels and decreases inflammation is a promising approach for prevention and treatment of atherosclerosis. In previous work, we reported that helper-dependent adenoviral (HDAd) overexpression of apolipoprotein A-I (apoAI) in endothelial cells (ECs) increases cholesterol efflux in vitro and reduces atherosclerosis in vivo. However, the effect of HDAdApoAI on atherosclerosis is partial. To improve this therapy, we considered concurrent overexpression of ATP-binding cassette subfamily A, member 1 (ABCA1), a protein that is required for apoAI-mediated cholesterol efflux. Before attempting combined apoAI/ABCA1 gene therapy, we tested whether an HDAd that expresses ABCA1 (HDAdABCA1) increases EC cholesterol efflux, whether increased cholesterol efflux alters normal EC physiology, and whether ABCA1 overexpression in ECs has anti-inflammatory effects. HDAdABCA1 increased EC ABCA1 protein (∼3-fold; p < 0.001) and apoAI-mediated cholesterol efflux (2.3-fold; p = 0.007). Under basal culture conditions, ABCA1 overexpression did not alter EC proliferation, metabolism, migration, apoptosis, nitric oxide production, or inflammatory gene expression. However, in serum-starved, apoAI-treated EC, ABCA1 overexpression had anti-inflammatory effects: decreased inflammatory gene expression (∼50%; p ≤ 0.02 for interleukin [IL]-6, tumor necrosis factor [TNF]-α, and vascular cell adhesion protein-1); reduced lipid-raft Toll-like receptor 4 (80%; p = 0.001); and a trend towards increased nitric oxide production (∼55%; p = 0.1). In ECs stimulated with lipopolysaccharide, ABCA1 overexpression markedly decreased inflammatory gene expression (∼90% for IL-6 and TNF-α; p < 0.001). Therefore, EC ABCA1 overexpression has no toxic effects and counteracts the two key drivers of atherosclerosis: cholesterol accumulation and inflammation. In vivo testing of HDAdABCA1 is warranted.
Collapse
Affiliation(s)
- Alexis Stamatikos
- 1 Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - Nagadhara Dronadula
- 1 Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - Philip Ng
- 2 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Donna Palmer
- 2 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Ethan Knight
- 1 Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - Bradley K Wacker
- 1 Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - Chongren Tang
- 1 Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - Francis Kim
- 1 Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - David A Dichek
- 1 Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
143
|
Gulec-Yilmaz S, Gulec H, Ogut DB, Cetin B, Gormus U, Isbir T. Association between apolipoprotein E genotypes and panic disorder in Turkish population. Nord J Psychiatry 2018; 72:477-483. [PMID: 29888635 DOI: 10.1080/08039488.2018.1482957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
AIM In this study, we aimed to investigate possible interactions among the apolipoprotein E (ApoE) and panic disorder (PD), taking into account serum cholesterol levels and subfractions. METHODS ApoE genotyping was performed by real-time polymerase chain reaction in DNA samples of PD patient group (n = 45) and healthy control group (n = 50). The serum lipid levels, low-density lipoprotein (LDL), and high-density lipoprotein (HDL) subfraction analysis were examined. RESULTS There was a significant difference of ApoE genotypes in patient and control groups. The E3/E3 genotypes lower whereas E4 allele carriers were significantly higher in PD group ApoE4allele carriers had 3.2-fold higher risk of PD. PD group had significantly lower LDL and HDL levels. In spite of the decreased levels of total LDL, antiatherogenic large LDL subgroup was significantly lower in a patient with PD. Antiatherogenic large HDL and Intermediate HDL levels were lower, while atherogenic small HDL subfraction was significantly higher in PD group. Furthermore, Apo E3/E3 genotype carriers had significantly higher large LDL, HDL, large HDL, intermediate HDL level, and also had highest HDL between all the groups. ApoE4 allele carriers while they had highest atherogenic small HDL level. CONCLUSION E4 allele can be associated with PD as an eligible risk factor, the E3/E3 could be a risk-reducing factor for PD. Patients with PD not only had lower LDL and HDL levels but also they have higher atherogenic LDL and HDL subfractions. Also, E3/E3 genotype carriers had convenient but ApoE4 carriers had atherogenic plasma cholesterol levels and subfractions.
Collapse
Affiliation(s)
- Seda Gulec-Yilmaz
- a Department of Molecular Medicine, Institute of Health Sciences , Yeditepe University , Istanbul , Turkey
| | - Huseyin Gulec
- b Erenkoy Mental and Neurological Disease Training and Research Hospital , University of Health Sciences , Istanbul , Turkey
| | - Dicle Bilge Ogut
- b Erenkoy Mental and Neurological Disease Training and Research Hospital , University of Health Sciences , Istanbul , Turkey
| | - Bugra Cetin
- c Department of Psychiatry, Faculty of Medicine , Maltepe University , Istanbul , Turkey
| | - Uzay Gormus
- d Department of Medical Biochemistry and Biophysics , Karolinska Institutet , Stockholm , Sweden
| | - Turgay Isbir
- e Department of Medical Biology, Faculty of Medicine , Yeditepe University , Istanbul , Turkey
| |
Collapse
|
144
|
Santos HO, Macedo RC. Cocoa-induced (Theobroma cacao) effects on cardiovascular system: HDL modulation pathways. Clin Nutr ESPEN 2018; 27:10-15. [DOI: 10.1016/j.clnesp.2018.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/10/2018] [Indexed: 02/05/2023]
|
145
|
Ling W, Li S, Zhang X, Xu Y, Gao Y, Du Q, Wang G, Fan W, Sun K, Bian J. Evaluation of Anti-Obesity Activity, Acute Toxicity, and Subacute Toxicity of Probiotic Dark Tea. Biomolecules 2018; 8:biom8040099. [PMID: 30257523 PMCID: PMC6316303 DOI: 10.3390/biom8040099] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/19/2018] [Accepted: 09/20/2018] [Indexed: 12/13/2022] Open
Abstract
Probiotic dark tea (PDT) is a novel kind of dark tea produced by fresh albino tea leaves and fermented with specific probiotics. Our study demonstrates that PDT can ameliorate high-fat diet-induced overweight and lipid metabolic disorders and shows no acute or subacute toxicity in Sprague-Dawley (SD) rats. Daily intragastric administration of 5% PDT infusion for 14 days caused no obvious effect on general physiological features and behaviors of rats. Oral administration of 1%, 2%, and 3% of PDT infusion for six weeks had no influence on the biochemistry and histopathology of rats’ organs and blood, as well as the body weight and ratios of organ/body weight. To investigate its anti-obesity activity, SD rats were randomly divided into four groups, treated with normal diet + water (Group I), high-fat diet + water (Group II), high-fat diet + 3% traditional dark tea infusion (Group III), high-fat diet + 3% PDT infusion (Group IV). After six weeks, the body weight, serum total triacylglycerol (TG) and serum total cholesterol (TC) levels of rats in Group II were significantly increased and the high-density lipoprotein cholesterol (HDL) levels were significantly decreased compared with those in the other three groups. Both traditional dark tea and PDT treatment effectively counteracted the adverse effect of a high-fat diet in SD rats. These results suggest that PDT could be applied for the prevention of obesity, which ameliorates overweight and lipid metabolic disorders and which shows no acute or subacute toxicity.
Collapse
Affiliation(s)
- Wang Ling
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-innovation Canter for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China.
| | - Shungeng Li
- Tianmu Lake Longxin Agricultural Ecological Park in Liyang City of Jiangsu Province, Liyang 213334, China.
| | - Xingcai Zhang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
| | - Yongquan Xu
- Tea Research Institute, Chinese Academy of Agricultural Sciences, 9 South Meiling Road, Hangzhou 310008, China.
| | - Ying Gao
- Tea Research Institute, Chinese Academy of Agricultural Sciences, 9 South Meiling Road, Hangzhou 310008, China.
| | - Qizhen Du
- College of Agricultural and Food Sciences, Zhejiang A & F University, Linan 311300, China.
| | - Guangguang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-innovation Canter for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China.
| | - Wentong Fan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-innovation Canter for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China.
| | - Kai Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-innovation Canter for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China.
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-innovation Canter for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China.
| |
Collapse
|
146
|
Kosmas CE, Silverio D, Sourlas A, Garcia F, Montan PD, Guzman E. Primary genetic disorders affecting high density lipoprotein (HDL). Drugs Context 2018; 7:212546. [PMID: 30214464 PMCID: PMC6135231 DOI: 10.7573/dic.212546] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 01/21/2023] Open
Abstract
There is extensive evidence demonstrating that there is a clear inverse correlation between plasma high density lipoprotein cholesterol (HDL-C) concentration and cardiovascular disease (CVD). On the other hand, there is also extensive evidence that HDL functionality plays a very important role in atheroprotection. Thus, genetic disorders altering certain enzymes, lipid transfer proteins, or specific receptors crucial for the metabolism and adequate function of HDL, may positively or negatively affect the HDL-C levels and/or HDL functionality and subsequently either provide protection or predispose to atherosclerotic disease. This review aims to describe certain genetic disorders associated with either low or high plasma HDL-C and discuss their clinical features, associated risk for cardiovascular events, and treatment options.
Collapse
Affiliation(s)
- Constantine E Kosmas
- Division of Cardiology, Department of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Delia Silverio
- Cardiology Clinic, Cardiology Unlimited, PC, New York, NY, USA
| | | | - Frank Garcia
- Cardiology Clinic, Cardiology Unlimited, PC, New York, NY, USA
| | - Peter D Montan
- Cardiology Clinic, Cardiology Unlimited, PC, New York, NY, USA
| | - Eliscer Guzman
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Bronx, NY, USA
| |
Collapse
|
147
|
Relationships of oxidized HDL with blood coagulation and fibrinolysis in patients with type 2 diabetes mellitus. J Thromb Thrombolysis 2018; 45:200-205. [PMID: 29247447 DOI: 10.1007/s11239-017-1594-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Although oxidization of LDL is known to be a crucial step for atherosclerotic progression, the significance of oxidized HDL remains to be clarified. The purpose of this study was to determine the relationships of oxidized HDL with blood coagulation and fibrinolysis in patients with diabetes. The subjects were outpatients with type 2 diabetes (n = 163; median hemoglobin A1c, 6.9%). Activities of blood coagulation and fibrinolysis were evaluated by levels of thrombin-anti-thrombin complex (TAT) and plasmin-α2 plasmin inhibitor complex (PIC), respectively. Relationships of oxidized HDL with TAT and PIC were investigated by using linear regression analysis and logistic regression analysis. Oxidized HDL showed a significant inverse correlation with TAT and a marginally significant correlation with PIC (Spearman's rank correlation coefficient: TAT, - 0.205 [p < 0.01]; PIC, - 0.135 [p = 0.087]). Prevalence of high TAT was significantly lower in the 3rd tertile group for oxidized HDL than in its 1st tertile (20.4 vs. 5.6%, p < 0.05), and prevalence of high PIC was marginally significantly lower in the 3rd tertile group for oxidized HDL than in its 1st tertile (40.7 vs. 24.1%, p = 0.099). In multivariate logistic regression analysis using age, gender, smoking, alcohol drinking, BMI, hemoglobin A1c, therapy for dyslipidemia, therapy for diabetes and anti-coagulation therapy as explanatory variables, odds ratios for high TAT and high PIC in the 3rd tertile group for oxidized HDL versus its 1st tertile group were significantly lower than the reference level of 1.00 (high TAT: 0.19 [0.04-0.99], p < 0.05; high PIC: 0.33 [0.12-0.95], p < 0.05). The frequency of high TAT or high PIC was lower in the higher tertile group for oxidized HDL than in its lower tertile group. Thus, oxidized HDL is thought to be inversely associated with both blood coagulation and fibrinolysis in patients with type 2 diabetes.
Collapse
|
148
|
Tall AR. Plasma high density lipoproteins: Therapeutic targeting and links to atherogenic inflammation. Atherosclerosis 2018; 276:39-43. [DOI: 10.1016/j.atherosclerosis.2018.07.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/19/2018] [Accepted: 07/03/2018] [Indexed: 11/16/2022]
|
149
|
Gao R, Liang JH, Wang L, Zhu HY, Wu W, Cao L, Fan L, Li JY, Yang T, Xu W. Low serum cholesterol levels predict inferior prognosis and improve NCCN-IPI scoring in diffuse large B cell lymphoma. Int J Cancer 2018; 143:1884-1895. [PMID: 29744861 DOI: 10.1002/ijc.31590] [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] [Received: 02/15/2018] [Revised: 04/15/2018] [Accepted: 04/23/2018] [Indexed: 01/08/2023]
Abstract
Low circulating cholesterol concentration is associated with elevated cancer incidence and mortality. However, the association between cholesterol levels and diffuse large B cell lymphoma (DLBCL) remains unknown. The aim of our study was to evaluate the prognostic value of serum lipid profile in DLBCL. Five hundred and fifty enrolled subjects with detailed serum lipid levels at diagnosis of DLBCL were randomly divided into a training set (n = 367) and a validation set (n = 183) (ratio, 2:1). Multivariate Cox regression analyses screened the prognostic factors associated with progression-free survival (PFS) and overall survival (OS). Performances of models were compared using C-index and area under the curve in internal and external validation. The results showed that decreased levels of total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C) were associated with unfavorable PFS and OS in the rituximab era, and concurrently low HDL-C together with low LDL-C was an independent prognostic indicator for both PFS and OS. Patients achieving complete remission or partial remission after 6-8 circles of chemotherapies had significantly increased cholesterol levels compared to the levels at DLBCL diagnosis, and HDL-C or LDL-C elevations were correlated with better survival. Furthermore, the predictive and discriminatory capacity of the National Comprehensive Cancer Network (NCCN)-International Prognostic Index (IPI) together with low cholesterol levels was superior to NCCN-IPI alone both in the training and validation set. In conclusion, serum cholesterol levels are simple and routinely tested parameters, which may be good candidates for predicting prognosis in the future clinical practice of DLBCL.
Collapse
Affiliation(s)
- Rui Gao
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China.,Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Jin-Hua Liang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Hua-Yuan Zhu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Wei Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Lei Cao
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Lei Fan
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Jian-Yong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Tao Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Wei Xu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| |
Collapse
|
150
|
Abstract
Foam cells are lipid-loaded macrophages and neutrophils that are generated from a massive uptake of oxidized lipid. Foam cells are a pathological hallmark of atherosclerosis, and have also been found in acne lesions. The same pathological changes determine the common pathogenesis. According to the pathological function of foam cells in these lesions, we put forward a viewpoint on the pathogenesis of acne and atherosclerotic plaques.
Collapse
|