1
|
Nicholls SJ, Tan S, Butters J, Nelson AJ. Evaluating obicetrapib as an emerging treatment for patients with dyslipidemia: a game changer? Expert Opin Pharmacother 2024; 25:1879-1885. [PMID: 39323412 DOI: 10.1080/14656566.2024.2409324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION Cholesteryl ester transfer protein (CETP) plays an important role in lipid metabolism. Early interest in the development of CETP inhibitors proved to be disappointing. Recent interest has focused on the potential for CETP inhibition to reduce cardiovascular risk by lowering levels of low-density lipoprotein cholesterol (LDL-C). AREAS COVERED The data suggesting that low CETP activity may associate with lower levels of cardiovascular risk and early experience with CETP inhibitors focused on raising HDL-C levels. More recent data that suggests that any potential to reduce cardiovascular risk by inhibition of CETP is more likely to result from lowering levels of atherogenic lipid parameters. The development of obicetrapib, a potent CETP inhibitor, with robust lowering of apoB and LDL-C, will be summarized as a potential approach to the prevention of cardiovascular disease. EXPERT OPINION Obicetrapib is a potent CETP inhibitor, with a demonstrated ability to lower levels of apoB and LDL-C as monotherapy and in addition to high intensity statin therapy. The ultimate impact of obicetrapib on cardiovascular events will be evaluated by ongoing clinical trials.
Collapse
Affiliation(s)
| | - Sean Tan
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Julie Butters
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Adam J Nelson
- Victorian Heart Institute, Monash University, Melbourne, Australia
| |
Collapse
|
2
|
Sheth S, Banach M, Toth PP. Closing the gap between guidelines and clinical practice for managing dyslipidemia: where are we now? Expert Rev Cardiovasc Ther 2024; 22:441-457. [PMID: 39198976 DOI: 10.1080/14779072.2024.2398444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 08/04/2024] [Accepted: 08/27/2024] [Indexed: 09/01/2024]
Abstract
INTRODUCTION Despite decades of research clearly illustrating the direct link between low-density lipoprotein cholesterol (LDL-C) and atherosclerotic cardiovascular disease (ASCVD) risk, LDL-C goal attainment rates are remarkably low in both the primary and secondary prevention settings. AREAS COVERED Herein we detail: (1) the low rates of LDL-C goal attainment; (2) despite guidelines clearly outlining indications of use, there is suboptimal initiation, intensification, and persistence of lipid lowering therapy, especially combination therapy; (3) key clinician-related factors contributing to this gap include inconsistent risk assessments, clinical inertia, and barriers to health access; (4) LDL-C reduction is associated with reductions in risk for cardiovascular events. Increasing LDL-C goal attainment rates should be a high public health priority. EXPERT OPINION There is an urgent need to rethink dyslipidemia management. Opportunities exist to overcome LDL-C goal attainment barriers, which necessitates a concerted effort from patients, clinicians, health systems, payors, pharmaceutical companies, and public health advocates. LDL-C measurement should be a performance metric for health systems. In addition, upfront use of combination therapy and polypill formulations should be encouraged. Engaging pharmacists to support drug therapy and adherence is crucial. Leveraging telehealth and electronic medical record (EMR) functionalities can enhance these efforts and ensure more effective implementation.
Collapse
Affiliation(s)
- Sohum Sheth
- University of Florida College of Medicine, Gainesville, FL, USA
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, Lodz, Poland
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter P Toth
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Preventive Cardiology, CGH Medical Center, Sterling, IL, USA
| |
Collapse
|
3
|
Nicholls SJ, Nelson AJ, Ditmarsch M, Kastelein JJP, Ballantyne CM, Ray KK, Navar AM, Nissen SE, Golberg AC, Brunham LR, Curcio D, Wuerdeman E, Neild A, Kling D, Hsieh A, Dicklin MR, Ference BA, Laufs U, Banach M, Mehran R, Catapano AL, Davidson MH. Obicetrapib on top of maximally tolerated lipid-modifying therapies in participants with or at high risk for atherosclerotic cardiovascular disease: rationale and designs of BROADWAY and BROOKLYN. Am Heart J 2024; 274:32-45. [PMID: 38705341 DOI: 10.1016/j.ahj.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/01/2024] [Accepted: 05/01/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Obicetrapib, a novel, selective cholesteryl ester transfer protein (CETP) inhibitor, reduces low-density lipoprotein cholesterol (LDL-C), LDL particles, apolipoprotein (Apo) B, and lipoprotein(a) [Lp(a)] and increases high-density lipoprotein cholesterol (HDL-C) when added to statins with or without ezetimibe. By substantially reducing LDL-C, obicetrapib has the potential to lower atherogenic lipoproteins in patients with atherosclerotic cardiovascular disease (ASCVD) or heterozygous familial hypercholesterolemia (HeFH) whose LDL-C levels remain high despite treatment with available maximally tolerated lipid-modifying therapies, addressing an unmet medical need in a patient population at high risk for cardiovascular events. METHODS AND RESULTS BROADWAY (NCT05142722) and BROOKLYN (NCT05425745) are ongoing placebo-controlled, double-blind, randomized Phase III trials designed to examine the efficacy, safety, and tolerability of obicetrapib as an adjunct to dietary intervention and maximally tolerated lipid-modifying therapies in participants with a history of ASCVD and/or underlying HeFH whose LDL-C is not adequately controlled. The primary efficacy endpoint was the percent change in LDL-C from baseline to day 84. Other endpoints included changes in Apo B, non-HDL-C, HDL-C, Apo A1, Lp(a), and triglycerides in addition to parameters evaluating safety, tolerability, and pharmacokinetics. BROADWAY also included an adjudicated assessment of major adverse cardiovascular events, measurements of glucose homeostasis, and an ambulatory blood pressure monitoring substudy. A total of 2,532 participants were randomized in BROADWAY and 354 in BROOKLYN to receive obicetrapib 10 mg or placebo (2:1) for 365 days with follow-up through 35 days after the last dose. Results from both trials are anticipated in 2024. CONCLUSION These trials will provide safety and efficacy data to support the potential use of obicetrapib among patients with ASCVD or HeFH with elevated LDL-C for whom existing therapies are not sufficiently effective or well-tolerated.
Collapse
Affiliation(s)
| | - Adam J Nelson
- Victorian Heart Institute, Monash University, Victoria, Australia
| | | | | | | | - Kausik K Ray
- Department of Primary Care and Public Health, Imperial College London, London, UK
| | | | - Steven E Nissen
- Cleveland Clinic Lerner School of Medicine at Case Western Reserve University, Cleveland, OH
| | | | - Liam R Brunham
- UBC Centre for Heart Lung Innovation, The University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | - Annie Neild
- NewAmsterdam Pharma, Naarden, The Netherlands
| | | | | | | | - Brian A Ference
- Centre for Naturally Randomized Trials, University of Cambridge, Cambridge, UK
| | - Ulrich Laufs
- Klinik und Poliklinkk für Kardiologie, Leipzig University, Germany
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidologym, Medical University of Lodz (MUL), Lodz, Poland
| | - Roxana Mehran
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alberico L Catapano
- IRCCS MultiMedica, Milan, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | | |
Collapse
|
4
|
Rehman WU, Yarkoni M, Ilyas MA, Athar F, Javaid M, Ehsan M, Khalid MT, Pasha A, Selma AB, Yarkoni A, Patel K, Sabouni MA, Rehman AU. Cholesteryl Ester Transfer Protein Inhibitors and Cardiovascular Outcomes: A Systematic Review and Meta-Analysis. J Cardiovasc Dev Dis 2024; 11:152. [PMID: 38786974 PMCID: PMC11122262 DOI: 10.3390/jcdd11050152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Atherosclerosis is a multi-factorial disease, and low-density lipoprotein cholesterol (LDL-C) is a critical risk factor in developing atherosclerotic cardiovascular disease (ASCVD). Cholesteryl-ester transfer-protein (CETP), synthesized by the liver, regulates LDL-C and high-density lipoprotein cholesterol (HDL-C) through the bidirectional transfer of lipids. The novelty of CETP inhibitors (CETPis) has granted new focus towards increasing HDL-C, besides lowering LDL-C strategies. To date, five CETPis that are projected to improve lipid profiles, torcetrapib, dalcetrapib, evacetrapib, anacetrapib, and obicetrapib, have reached late-stage clinical development for ASCVD risk reduction. Early trials failed to reduce atherosclerotic cardiovascular occurrences. Given the advent of some recent large-scale clinical trials (ACCELERATE, HPS3/TIMI55-REVEAL Collaborative Group), conducting a meta-analysis is essential to investigate CETPis' efficacy. METHODS We conducted a thorough search of randomized controlled trials (RCTs) that commenced between 2003 and 2023; CETPi versus placebo studies with a ≥6-month follow-up and defined outcomes were eligible. PRIMARY OUTCOMES major adverse cardiovascular events (MACEs), cardiovascular disease (CVD)-related mortality, all-cause mortality. SECONDARY OUTCOMES stroke, revascularization, hospitalization due to acute coronary syndrome, myocardial infarction (MI). RESULTS Nine RCTs revealed that the use of a CETPi significantly reduced CVD-related mortality (RR = 0.89; 95% CI: 0.81-0.98; p = 0.02; I2 = 0%); the same studies also reduced the risk of MI (RR = 0.92; 95% CI: 0.86-0.98; p = 0.01; I2 = 0%), which was primarily attributed to anacetrapib. The use of a CETPi did not reduce the likelihood any other outcomes. CONCLUSIONS Our meta-analysis shows, for the first time, that CETPis are associated with reduced CVD-related mortality and MI.
Collapse
Affiliation(s)
- Wajeeh ur Rehman
- Heart and Vascular Institute, United Health Services, Johnson City, NY 13790, USA; (A.P.); (A.Y.); (K.P.); (A.u.R.)
| | - Merav Yarkoni
- Heart and Vascular Institute, United Health Services, Johnson City, NY 13790, USA; (A.P.); (A.Y.); (K.P.); (A.u.R.)
| | - Muhammad Abdullah Ilyas
- Department of Medicine, King Edward Medical University, Lahore 54000, Pakistan; (M.A.I.); (F.A.); (M.E.)
| | - Farwa Athar
- Department of Medicine, King Edward Medical University, Lahore 54000, Pakistan; (M.A.I.); (F.A.); (M.E.)
| | - Mahnoor Javaid
- School of Medicine, CMH Lahore Medical College, Lahore 54000, Pakistan;
| | - Muhammad Ehsan
- Department of Medicine, King Edward Medical University, Lahore 54000, Pakistan; (M.A.I.); (F.A.); (M.E.)
| | - Muhammad Talha Khalid
- Department of Medicine, United Health Services, Johnson City, NY 13790, USA; (M.T.K.); (A.B.S.)
| | - Ahmed Pasha
- Heart and Vascular Institute, United Health Services, Johnson City, NY 13790, USA; (A.P.); (A.Y.); (K.P.); (A.u.R.)
| | - Abdelhamid Ben Selma
- Department of Medicine, United Health Services, Johnson City, NY 13790, USA; (M.T.K.); (A.B.S.)
| | - Alon Yarkoni
- Heart and Vascular Institute, United Health Services, Johnson City, NY 13790, USA; (A.P.); (A.Y.); (K.P.); (A.u.R.)
| | - Keyoor Patel
- Heart and Vascular Institute, United Health Services, Johnson City, NY 13790, USA; (A.P.); (A.Y.); (K.P.); (A.u.R.)
| | - Mouhamed Amr Sabouni
- Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Afzal ur Rehman
- Heart and Vascular Institute, United Health Services, Johnson City, NY 13790, USA; (A.P.); (A.Y.); (K.P.); (A.u.R.)
| |
Collapse
|
5
|
Agnello F, Ingala S, Laterra G, Scalia L, Barbanti M. Novel and Emerging LDL-C Lowering Strategies: A New Era of Dyslipidemia Management. J Clin Med 2024; 13:1251. [PMID: 38592091 PMCID: PMC10931739 DOI: 10.3390/jcm13051251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/09/2024] [Accepted: 02/20/2024] [Indexed: 04/10/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) represents a major global health challenge, significantly contributing to mortality rates. This chronic inflammatory condition affecting blood vessels is intricately linked to hypercholesterolemia, with elevated levels of low-density lipoprotein cholesterol (LDL-C) recognized as a central and modifiable risk factor. The effectiveness of lipid-lowering therapy (LLT) in mitigating ASCVD risk is well established, with studies revealing a substantial reduction in major ischemic events correlating with LDL-C reduction. While statins, often combined with ezetimibe, remain fundamental in dyslipidemia management, a significant proportion of patients on statin therapy continue to experience cardiovascular events. Recent pharmacological advancements, driven by a deeper understanding of atherogenesis, have unveiled novel therapeutic targets and potent drugs. Notably, agents like bempedoic acid and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors (evolocumab, alirocumab, inclisiran) have emerged as effective options to intensify LLT and achieve LDL-C goals, addressing limitations associated with statins, such as myopathy. Molecular insights into alternative pathways have spurred the investigation of emerging agents, offering promising perspectives for novel medications with efficacy comparable to established treatments, associated with advantages in cost and administration. This review provides a comprehensive overview of the evolving landscape of lipid-lowering strategies, highlighting the progress made in addressing ASCVD risk and the potential of upcoming therapies to further optimize cardiovascular prevention.
Collapse
Affiliation(s)
- Federica Agnello
- Division of Cardiology, Ospedale Umberto I, ASP 4 di Enna, 94100 Enna, Italy; (F.A.); (S.I.); (L.S.)
| | - Salvatore Ingala
- Division of Cardiology, Ospedale Umberto I, ASP 4 di Enna, 94100 Enna, Italy; (F.A.); (S.I.); (L.S.)
| | - Giulia Laterra
- Division of Cardiology, Ospedale Umberto I, ASP 4 di Enna, 94100 Enna, Italy; (F.A.); (S.I.); (L.S.)
| | - Lorenzo Scalia
- Division of Cardiology, Ospedale Umberto I, ASP 4 di Enna, 94100 Enna, Italy; (F.A.); (S.I.); (L.S.)
| | - Marco Barbanti
- Division of Cardiology, Ospedale Umberto I, ASP 4 di Enna, 94100 Enna, Italy; (F.A.); (S.I.); (L.S.)
- Faculty of Medicine and Surgery, Università degli Studi di Enna “Kore”, 94100 Enna, Italy
| |
Collapse
|
6
|
Ølnes ÅS, Teigen M, Laerdahl JK, Leren TP, Strøm TB, Bjune K. Variants in the CETP gene affect levels of HDL cholesterol by reducing the amount, and not the specific lipid transfer activity, of secreted CETP. PLoS One 2023; 18:e0294764. [PMID: 38039300 PMCID: PMC10691695 DOI: 10.1371/journal.pone.0294764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/31/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Cholesteryl ester transfer protein (CETP) transfers cholesteryl esters in plasma from high density lipoprotein (HDL) to very low density lipoprotein and low density lipoprotein. Loss-of-function variants in the CETP gene cause elevated levels of HDL cholesterol. In this study, we have determined the functional consequences of 24 missense variants in the CETP gene. The 24 missense variants studied were the ones reported in the Human Gene Mutation Database and in the literature to affect HDL cholesterol levels, as well as two novel variants identified at the Unit for Cardiac and Cardiovascular Genetics, Oslo University Hospital in subjects with hyperalphalipoproteinemia. METHODS HEK293 cells were transiently transfected with mutant CETP plasmids. The amounts of CETP protein in lysates and media were determined by Western blot analysis, and the lipid transfer activities of the CETP variants were determined by a fluorescence-based assay. RESULTS Four of the CETP variants were not secreted. Five of the variants were secreted less than 15% compared to the WT-CETP, while the other 15 variants were secreted in varying amounts. There was a linear relationship between the levels of secreted protein and the lipid transfer activities (r = 0.96, p<0.001). Thus, the secreted variants had similar specific lipid transfer activities. CONCLUSION The effect of the 24 missense variants in the CETP gene on the lipid transfer activity was mediated predominantly by their impact on the secretion of the CETP protein. The four variants that prevented CETP secretion cause autosomal dominant hyperalphalipoproteinemia. The five variants that markedly reduced secretion of the respective variants cause mild hyperalphalipoproteinemia. The majority of the remaining 15 variants had minor effects on the secretion of CETP, and are considered neutral genetic variants.
Collapse
Affiliation(s)
- Åsa Schawlann Ølnes
- Unit for Cardiac and Cardiovascular Genetics, Oslo University Hospital, Oslo, Norway
| | - Marianne Teigen
- Unit for Cardiac and Cardiovascular Genetics, Oslo University Hospital, Oslo, Norway
| | - Jon K. Laerdahl
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
- Department of Informatics, ELIXIR Norway, University of Oslo, Oslo, Norway
| | - Trond P. Leren
- Unit for Cardiac and Cardiovascular Genetics, Oslo University Hospital, Oslo, Norway
| | - Thea Bismo Strøm
- Unit for Cardiac and Cardiovascular Genetics, Oslo University Hospital, Oslo, Norway
| | - Katrine Bjune
- Unit for Cardiac and Cardiovascular Genetics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
7
|
Mehta N, Dangas K, Ditmarsch M, Rensen PCN, Dicklin MR, Kastelein JJP. The evolving role of cholesteryl ester transfer protein inhibition beyond cardiovascular disease. Pharmacol Res 2023; 197:106972. [PMID: 37898443 DOI: 10.1016/j.phrs.2023.106972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 09/21/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
The main role of cholesteryl ester transfer protein (CETP) is the transfer of cholesteryl esters and triglycerides between high-density lipoprotein (HDL) particles and triglyceride-rich lipoprotein and low-density lipoprotein (LDL) particles. There is a long history of investigations regarding the inhibition of CETP as a target for reducing major adverse cardiovascular events. Initially, the potential effect on cardiovascular events of CETP inhibitors was hypothesized to be mediated by their ability to increase HDL cholesterol, but, based on evidence from anacetrapib and the newest CETP inhibitor, obicetrapib, it is now understood to be primarily due to reducing LDL cholesterol and apolipoprotein B. Nevertheless, evidence is also mounting that other roles of HDL, including its promotion of cholesterol efflux, as well as its apolipoprotein composition and anti-inflammatory, anti-oxidative, and anti-diabetic properties, may play important roles in several diseases beyond cardiovascular disease, including, but not limited to, Alzheimer's disease, diabetes, and sepsis. Furthermore, although Mendelian randomization analyses suggested that higher HDL cholesterol is associated with increased risk of age-related macular degeneration (AMD), excess risk of AMD was absent in all CETP inhibitor randomized controlled trial data comprising over 70,000 patients. In fact, certain HDL subclasses may, in contrast, be beneficial for treating the retinal cholesterol accumulation that occurs with AMD. This review describes the latest biological evidence regarding the relationship between HDL and CETP inhibition for Alzheimer's disease, type 2 diabetes mellitus, sepsis, and AMD.
Collapse
Affiliation(s)
- Nehal Mehta
- Mobius Scientific, Inc., JLABS @ Washington, DC, Washington, DC, USA
| | | | | | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory of Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | | | - John J P Kastelein
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Mohamed F, Mansfield B, Raal FJ. Targeting PCSK9 and Beyond for the Management of Low-Density Lipoprotein Cholesterol. J Clin Med 2023; 12:5082. [PMID: 37568484 PMCID: PMC10419884 DOI: 10.3390/jcm12155082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Reducing low-density lipoprotein cholesterol (LDL-C) levels is crucial to the prevention of atherosclerotic cardiovascular disease (ASCVD). However, many patients, especially those at very high ASCVD risk or with familial hypercholesterolemia (FH), do not achieve target LDL-C levels with statin monotherapy. The underutilization of novel lipid-lowering therapies (LLT) globally may be due to cost concerns or therapeutic inertia. Emerging approaches have the potential to lower LDL-C and reduce ASCVD risk further, in addition to offering alternatives for statin-intolerant patients. Shifting the treatment paradigm towards initial combination therapy and utilizing novel LLT strategies can complement existing treatments. This review discusses innovative approaches including combination therapies involving statins and agents like ezetimibe, bempedoic acid, cholesterol ester transfer protein (CETP) inhibitors as well as strategies targeting proprotein convertase subtilisin/kexin type 9 (PCSK9) and angiopoietin-like protein 3 (ANGPTL3) inhibition. Advances in nucleic acid-based therapies and gene editing are innovative approaches that will improve patient compliance and adherence. These strategies demonstrate significant LDL-C reductions and improved cardiovascular outcomes, offering potential for optimal LDL-C control and reduced ASCVD risk. By addressing the limitations of statin monotherapy, these approaches provide new management options for elevated LDL-C levels.
Collapse
Affiliation(s)
| | | | - Frederick J. Raal
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa; (F.M.); (B.M.)
| |
Collapse
|
9
|
Nicholls SJ, Ray KK, Nelson AJ, Kastelein JJP. Can we revive CETP-inhibitors for the prevention of cardiovascular disease? Curr Opin Lipidol 2022; 33:319-325. [PMID: 36345867 DOI: 10.1097/mol.0000000000000854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE OF REVIEW To review recent developments in the field of cholesteryl ester transfer protein (CETP) inhibition from clinical trials and genomic analyses which have the potential to impact future clinical programs. RECENT FINDINGS CETP plays an important role in remodelling of lipoproteins. A large body of evidence suggests that the presence of low CETP activity should have favourable effects on lipid profiles and cardiovascular risk. However, a number of clinical development programs of pharmacological CETP inhibitors have been disappointing with reports of toxicity and clinical futility. These findings have led many to consider abandoning CETP inhibition as a potential strategy for cardiovascular prevention. However, recent observations from genomic analyses and post hoc observations of prior clinical trials have given greater insights into the potential relationship between CETP inhibition and cardiovascular risk. This has highlighted the importance of lowering levels of atherogenic lipoproteins. SUMMARY These findings provide a pathway for ongoing clinical development of CETP inhibitors, where the potential to play an important role in the prevention of cardiovascular disease may still be possible. The lessons learned and pathway forward for new CETP inhibitors will be reviewed.
Collapse
Affiliation(s)
| | | | - Adam J Nelson
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - John J P Kastelein
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Nelson AJ, Sniderman AD, Ditmarsch M, Dicklin MR, Nicholls SJ, Davidson MH, Kastelein JJP. Cholesteryl Ester Transfer Protein Inhibition Reduces Major Adverse Cardiovascular Events by Lowering Apolipoprotein B Levels. Int J Mol Sci 2022; 23:ijms23169417. [PMID: 36012684 PMCID: PMC9409323 DOI: 10.3390/ijms23169417] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 12/04/2022] Open
Abstract
Cholesteryl ester transfer protein (CETP) facilitates the exchange of cholesteryl esters and triglycerides (TG) between high-density lipoprotein (HDL) particles and TG-rich, apolipoprotein (apo) B-containing particles. Initially, these compounds were developed to raise plasma HDL cholesterol (HDL-C) levels, a mechanism that was previously thought to lower the risk of atherosclerotic cardiovascular disease (ASCVD). More recently, the focus changed and the use of pharmacologic CETP inhibitors to reduce low-density lipoprotein cholesterol (LDL-C), non-HDL-C and apoB concentrations became supported by several lines of evidence from animal models, observational investigations, randomized controlled trials and Mendelian randomization studies. Furthermore, a cardiovascular outcome trial of anacetrapib demonstrated that CETP inhibition significantly reduced the risk of major coronary events in patients with ASCVD in a manner directly proportional to the substantial reduction in LDL-C and apoB. These data have dramatically shifted the attention on CETP away from raising HDL-C instead to lowering apoB-containing lipoproteins, which is relevant since the newest CETP inhibitor, obicetrapib, reduces LDL-C by up to 51% and apoB by up to 30% when taken in combination with a high-intensity statin. An ongoing cardiovascular outcome trial of obicetrapib in patients with ASCVD is expected to provide further evidence of the ability of CETP inhibitors to reduce major adverse cardiovascular events by lowering apoB. The purpose of the present review is to provide an up-to-date understanding of CETP inhibition and its relationship to ASCVD risk reduction.
Collapse
Affiliation(s)
- Adam J. Nelson
- Victorian Heart Institute, Monash University, Clayton, VIC 3800, Australia
| | - Allan D. Sniderman
- Mike and Valeria Rosenbloom Centre for Cardiovascular Prevention, Department of Medicine, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | | | | | | | | | - John J. P. Kastelein
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Correspondence:
| |
Collapse
|
11
|
Association of lipid metabolism-related gene promoter methylation with risk of coronary artery disease. Mol Biol Rep 2022; 49:9373-9378. [PMID: 35941416 DOI: 10.1007/s11033-022-07789-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 10/15/2022]
Abstract
BACKGROUND Coronary artery disease (CAD) is a complex disease that is influenced by environmental and genetic factors. Lipid levels are regarded as a major risk factor for CAD, and epigenetic mechanisms might be involved in the regulation of CAD development. This study was designed to investigate the association between the DNA methylation status of 8 lipid metabolism-related genes and the risk of CAD in the Chinese Han population. METHODS A total of 260 individuals were sampled in this study, including 120 CAD cases and 140 normal healthy controls. DNA methylation status was tested via targeted bisulfite sequencing. RESULTS The results indicated a significant association between hypomethylation of the APOC3, CETP and APOC1 gene promoters and the risk of CAD. Individuals with higher methylation levels of the APOA5 and LIPC gene promoters had increased risks for CAD. In addition, ANGPTL4 methylation level was significantly associated with CAD in males but not females. There were no significant differences in the methylation levels of the APOB and PCSK9 gene promoters between CAD patients and controls. CONCLUSIONS The methylation status of the APOC3, APOA5, LIPC, CETP and APOC1 gene promoters may be associated with the development of CAD.
Collapse
|
12
|
Yokoyama S. HDL Receptor in Schistosoma japonicum Mediating Egg Embryonation: Potential Molecular Basis for High Prevalence of Cholesteryl Ester Transfer Protein Deficiency in East Asia. Front Cell Dev Biol 2022; 10:807289. [PMID: 35372338 PMCID: PMC8968628 DOI: 10.3389/fcell.2022.807289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/21/2022] [Indexed: 12/03/2022] Open
Abstract
Schistosomiasis is a life-threatening parasitic disease caused by blood flukes, Schistosomes. In its intestinal type, the parasites reside in visceral/portal veins of the human hosts and lay eggs to excrete in feces via intestinal tracts, and some of the aberrant eggs plug into the liver via the portal blood flow. Ectopic growth of these eggs causes fatal granulomatosis and cirrhosis of the liver. The parasites ingest nutrients from the host blood plasma by using nonspecific and specific transport via their body surface and alimentary tracts. It is especially important for the female adults to obtain lipid molecules because they synthesize neither fatty acids nor sterols and yet produce egg yolk. Low-density lipoprotein receptors have been identified in the body of the Schistosomes but their functions in the parasite life cycle have not clearly been characterized. On the other hand, CD36-related protein was identified in the body and the eggs of Asian blood fluke, Schistosoma japonicum, and characterized as a molecule that mediates selective uptake of cholesteryl ester from the host plasma high-density lipoproteins (HDLs). This reaction was shown crucial for their eggs to grow to miracidia. Interestingly, abnormal large HDL generated in lack of cholesteryl ester transfer protein (CETP) is a poor substrate for this reaction, and, therefore, CETP deficiency resists pathogenic ectopic growth of the aberrant parasite eggs in the liver. This genetic mutation is exclusively found in East Asia, overlapping with the current and historic regions of Schistosoma japonicum epidemic, so that this infection could be related to high prevalence of CETP deficiency in East Asia.
Collapse
Affiliation(s)
- Shinji Yokoyama
- Food and Nutritional Sciences, Chubu University, Kasugai, Japan
| |
Collapse
|
13
|
Nurmohamed NS, Ditmarsch M, Kastelein JJP. CETP-inhibitors: from HDL-C to LDL-C lowering agents? Cardiovasc Res 2021; 118:2919-2931. [PMID: 34849601 PMCID: PMC9648826 DOI: 10.1093/cvr/cvab350] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/20/2021] [Indexed: 11/29/2022] Open
Abstract
Cholesteryl ester transfer protein (CETP) is a liver-synthesized glycoprotein whose main functions are facilitating transfer of both cholesteryl esters from high-density lipoprotein (HDL) particles to apolipoprotein B (apoB)-containing particles as well as transfer of triglycerides from apoB-containing particles to HDL particles. Novel crystallographic data have shown that CETP exchanges lipids in the circulation by a dual molecular mechanism. Recently, it has been suggested that the atherosclerotic cardiovascular disease (ASCVD) benefit from CETP inhibition is the consequence of the achieved low-density lipoprotein cholesterol (LDL-C) and apoB reduction, rather than through the HDL cholesterol (HDL-C) increase. The use of CETP inhibitors is supported by genetic evidence from Mendelian randomization studies, showing that LDL-C lowering by CETP gene variants achieves equal ASCVD risk reduction as LDL-C lowering through gene proxies for statins, ezetimibe, and proprotein convertase subtilisin–kexin Type 9 inhibitors. Although first-generation CETP inhibitors (torcetrapib, dalcetrapib) were mainly raising HDL-C or had off-target effects, next generation CETP inhibitors (anacetrapib, evacetrapib) were also effective in reducing LDL-C and apoB and have been proven safe. Anacetrapib was the first CETP inhibitor to be proven effective in reducing ASCVD risk. In addition, CETP inhibitors have been shown to lower the risk of new-onset diabetes, improve glucose tolerance, and insulin sensitivity. The newest-generation CETP inhibitor obicetrapib, specifically designed to lower LDL-C and apoB, has achieved significant reductions of LDL-C up to 45%. Obicetrapib, about to enter phase III development, could become the first CETP inhibitor as add-on therapy for patients not reaching their guideline LDL-C targets.
Collapse
Affiliation(s)
- Nick S Nurmohamed
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Cardiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - John J P Kastelein
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Kardassis D, Thymiakou E, Chroni A. Genetics and regulation of HDL metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1867:159060. [PMID: 34624513 DOI: 10.1016/j.bbalip.2021.159060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023]
Abstract
The inverse association between plasma HDL cholesterol (HDL-C) levels and risk for cardiovascular disease (CVD) has been demonstrated by numerous epidemiological studies. However, efforts to reduce CVD risk by pharmaceutically manipulating HDL-C levels failed and refused the HDL hypothesis. HDL-C levels in the general population are highly heterogeneous and are determined by a combination of genetic and environmental factors. Insights into the causes of HDL-C heterogeneity came from the study of monogenic HDL deficiency syndromes but also from genome wide association and Μendelian randomization studies which revealed the contribution of a large number of loci to low or high HDL-C cases in the general or in restricted ethnic populations. Furthermore, HDL-C levels in the plasma are under the control of transcription factor families acting primarily in the liver including members of the hormone nuclear receptors (PPARs, LXRs, HNF-4) and forkhead box proteins (FOXO1-4) and activating transcription factors (ATFs). The effects of certain lipid lowering drugs used today are based on the modulation of the activity of specific members of these transcription factors. During the past decade, the roles of small or long non-coding RNAs acting post-transcriptionally on the expression of HDL genes have emerged and provided novel insights into HDL regulation and new opportunities for therapeutic interventions. In the present review we summarize recent progress made in the genetics and the regulation (transcriptional and post-transcriptional) of HDL metabolism.
Collapse
Affiliation(s)
- Dimitris Kardassis
- Laboratory of Biochemistry, Department of Basic Sciences, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion, Greece.
| | - Efstathia Thymiakou
- Laboratory of Biochemistry, Department of Basic Sciences, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion, Greece
| | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| |
Collapse
|
15
|
Giroud S, Chery I, Arrivé M, Prost M, Zumsteg J, Heintz D, Evans AL, Gauquelin-Koch G, Arnemo JM, Swenson JE, Lefai E, Bertile F, Simon C, Blanc S. Hibernating brown bears are protected against atherogenic dyslipidemia. Sci Rep 2021; 11:18723. [PMID: 34548543 PMCID: PMC8455566 DOI: 10.1038/s41598-021-98085-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022] Open
Abstract
To investigate mechanisms by which hibernators avoid atherogenic hyperlipidemia during hibernation, we assessed lipoprotein and cholesterol metabolisms of free-ranging Scandinavian brown bears (Ursus arctos). In winter- and summer-captured bears, we measured lipoprotein sizes and sub-classes, triglyceride-related plasma-enzyme activities, and muscle lipid composition along with plasma-levels of antioxidant capacities and inflammatory markers. Although hibernating bears increased nearly all lipid levels, a 36%-higher cholesteryl-ester transfer-protein activity allowed to stabilize lipid composition of high-density lipoproteins (HDL). Levels of inflammatory metabolites, i.e., 7-ketocholesterol and 11ß-prostaglandin F2α, declined in winter and correlated inversely with cardioprotective HDL2b-proportions and HDL-sizes that increased during hibernation. Lower muscle-cholesterol concentrations and lecithin-cholesterol acyltransferase activity in winter suggest that hibernating bears tightly controlled peripheral-cholesterol synthesis and/or release. Finally, greater plasma-antioxidant capacities prevented excessive lipid-specific oxidative damages in plasma and muscles of hibernating bears. Hence, the brown bear manages large lipid fluxes during hibernation, without developing adverse atherogenic effects that occur in humans and non-hibernators.
Collapse
Affiliation(s)
- Sylvain Giroud
- Research Institute of Wildlife Ecology, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Savoyenstraße 1, 1160, Vienna, Austria.
| | - Isabelle Chery
- University of Strasbourg, 4 rue Blaise Pascal, 67081, Strasbourg, France.,CNRS, UMR7178, Institut Pluridisciplinaire Hubert Curien (IPHC), 23 rue du Loess, 67087, Strasbourg, France
| | - Mathilde Arrivé
- University of Strasbourg, 4 rue Blaise Pascal, 67081, Strasbourg, France.,CNRS, UMR7178, Institut Pluridisciplinaire Hubert Curien (IPHC), 23 rue du Loess, 67087, Strasbourg, France
| | | | - Julie Zumsteg
- Plant Imaging & Mass Spectrometry (PIMS), Institute of Plant Molecular Biology, CNRS, University of Strasbourg, 12 rue du Général Zimmer, 67084, Strasbourg, France
| | - Dimitri Heintz
- Plant Imaging & Mass Spectrometry (PIMS), Institute of Plant Molecular Biology, CNRS, University of Strasbourg, 12 rue du Général Zimmer, 67084, Strasbourg, France
| | - Alina L Evans
- Department of Forestry and Wildlife Management, Inland Norway University of Applied Sciences, 2480, Koppang, Norway
| | | | - Jon M Arnemo
- Department of Forestry and Wildlife Management, Inland Norway University of Applied Sciences, 2480, Koppang, Norway.,Department of Wildlife, Fish and Environmental Studies, Swedish University of Agricultural Sciences, 90183, Umeå, Sweden
| | - Jon E Swenson
- Faculty of Environmental Sciences and Natural Resource Management, Norwegian University of Life Sciences, PO Box 5003, 1432, Ås, Norway
| | - Etienne Lefai
- University of Auvergne, INRAE, UNH UMR1019, 63122, Saint-Genès Champanelle, France
| | - Fabrice Bertile
- University of Strasbourg, 4 rue Blaise Pascal, 67081, Strasbourg, France.,CNRS, UMR7178, Institut Pluridisciplinaire Hubert Curien (IPHC), 23 rue du Loess, 67087, Strasbourg, France
| | - Chantal Simon
- CARMEN, INSERM U1060/University of Lyon / INRA U1235, Oullins, France
| | - Stéphane Blanc
- University of Strasbourg, 4 rue Blaise Pascal, 67081, Strasbourg, France.,CNRS, UMR7178, Institut Pluridisciplinaire Hubert Curien (IPHC), 23 rue du Loess, 67087, Strasbourg, France
| |
Collapse
|
16
|
Giammanco A, Noto D, Barbagallo CM, Nardi E, Caldarella R, Ciaccio M, Averna MR, Cefalù AB. Hyperalphalipoproteinemia and Beyond: The Role of HDL in Cardiovascular Diseases. Life (Basel) 2021; 11:life11060581. [PMID: 34207236 PMCID: PMC8235218 DOI: 10.3390/life11060581] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 12/16/2022] Open
Abstract
Hyperalphalipoproteinemia (HALP) is a lipid disorder characterized by elevated plasma high-density lipoprotein cholesterol (HDL-C) levels above the 90th percentile of the distribution of HDL-C values in the general population. Secondary non-genetic factors such as drugs, pregnancy, alcohol intake, and liver diseases might induce HDL increases. Primary forms of HALP are caused by mutations in the genes coding for cholesteryl ester transfer protein (CETP), hepatic lipase (HL), apolipoprotein C-III (apo C-III), scavenger receptor class B type I (SR-BI) and endothelial lipase (EL). However, in the last decades, genome-wide association studies (GWAS) have also suggested a polygenic inheritance of hyperalphalipoproteinemia. Epidemiological studies have suggested that HDL-C is inversely correlated with cardiovascular (CV) risk, but recent Mendelian randomization data have shown a lack of atheroprotective causal effects of HDL-C. This review will focus on primary forms of HALP, the role of polygenic inheritance on HDL-C, associated risk for cardiovascular diseases and possible treatment options.
Collapse
Affiliation(s)
- Antonina Giammanco
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties–University of Palermo, Via del Vespro, 129, 90127 Palermo, Italy; (A.G.); (D.N.); (C.M.B.); (E.N.); (M.R.A.)
| | - Davide Noto
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties–University of Palermo, Via del Vespro, 129, 90127 Palermo, Italy; (A.G.); (D.N.); (C.M.B.); (E.N.); (M.R.A.)
| | - Carlo Maria Barbagallo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties–University of Palermo, Via del Vespro, 129, 90127 Palermo, Italy; (A.G.); (D.N.); (C.M.B.); (E.N.); (M.R.A.)
| | - Emilio Nardi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties–University of Palermo, Via del Vespro, 129, 90127 Palermo, Italy; (A.G.); (D.N.); (C.M.B.); (E.N.); (M.R.A.)
| | - Rosalia Caldarella
- Department of Laboratory Medicine, Unit of Laboratory Medicine CoreLab, University Hospital “P. Giaccone”, 90127 Palermo, Italy; (R.C.); (M.C.)
| | - Marcello Ciaccio
- Department of Laboratory Medicine, Unit of Laboratory Medicine CoreLab, University Hospital “P. Giaccone”, 90127 Palermo, Italy; (R.C.); (M.C.)
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| | - Maurizio Rocco Averna
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties–University of Palermo, Via del Vespro, 129, 90127 Palermo, Italy; (A.G.); (D.N.); (C.M.B.); (E.N.); (M.R.A.)
| | - Angelo Baldassare Cefalù
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties–University of Palermo, Via del Vespro, 129, 90127 Palermo, Italy; (A.G.); (D.N.); (C.M.B.); (E.N.); (M.R.A.)
- Correspondence:
| |
Collapse
|
17
|
Park JH, Lee CW, Nam MJ, Kim H, Kwon DY, Yoo JW, Lee KN, Han K, Jung JH, Park YG, Kim DH. Association of High-Density Lipoprotein Cholesterol Variability and the Risk of Developing Parkinson Disease. Neurology 2021; 96:e1391-e1401. [PMID: 33536275 DOI: 10.1212/wnl.0000000000011553] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/09/2020] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To investigate the longitudinal association among high-density lipoprotein cholesterol (HDL-C) level, HDL-C variability, and the risk of developing Parkinson disease (PD). METHODS We conducted a nationwide, population-based cohort study. We included 382,391 patients aged ≥65 years who underwent at least 3 health examinations provided by the Korean National Health Insurance System from 2008 to 2013 and followed up until 2017. Individuals with a history of PD and missing values were excluded (n = 1,987). We assessed HDL-C variability using 3 indices, including variability independent of the mean (VIM). A multivariate-adjusted Cox proportional hazards regression analysis was performed. RESULTS Among the 380,404 participants, 2,733 individuals were newly diagnosed with PD during a median follow-up period of 5 years. The lowest quartile (Q1) group of baseline HDL-C and mean HDL-C was associated with increased PD incidence as compared with the highest quartile (Q4) group (adjusted hazard ratio [aHR], 1.20; 95% confidence interval [CI], 1.08-1.34; and aHR, 1.16; 95% CI, 1.04-1.30, respectively). The Q4 group of HDL-C variability (VIM) was associated with increased PD incidence compared to the Q1 group (aHR, 1.19; 95% CI, 1.06-1.33). The group with the Q1 of baseline HDL-C and with the Q4 of HDL-C variability had the highest risk of PD incidence (aHR, 1.6; 95% CI, 1.31-1.96). CONCLUSION Lower HDL-C level and greater HDL-C variability were associated with a higher incidence of PD.
Collapse
Affiliation(s)
- Joo-Hyun Park
- From the Departments of Family Medicine (J.-H.P., C.-w.L., M.J.N., H.K., D.-H.K.) and Neurology (D.-Y.K.), Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea; Department of Internal Medicine (J.W.Y.), University of Nevada Las Vegas School of Medicine; Department of Statistics and Actuarial Science (K.N.L., K.H.), Soongsil University; and Department of Biostatistics (J.-H.J., Y.-G.P.), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Chung-Woo Lee
- From the Departments of Family Medicine (J.-H.P., C.-w.L., M.J.N., H.K., D.-H.K.) and Neurology (D.-Y.K.), Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea; Department of Internal Medicine (J.W.Y.), University of Nevada Las Vegas School of Medicine; Department of Statistics and Actuarial Science (K.N.L., K.H.), Soongsil University; and Department of Biostatistics (J.-H.J., Y.-G.P.), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Myung Ji Nam
- From the Departments of Family Medicine (J.-H.P., C.-w.L., M.J.N., H.K., D.-H.K.) and Neurology (D.-Y.K.), Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea; Department of Internal Medicine (J.W.Y.), University of Nevada Las Vegas School of Medicine; Department of Statistics and Actuarial Science (K.N.L., K.H.), Soongsil University; and Department of Biostatistics (J.-H.J., Y.-G.P.), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyunjin Kim
- From the Departments of Family Medicine (J.-H.P., C.-w.L., M.J.N., H.K., D.-H.K.) and Neurology (D.-Y.K.), Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea; Department of Internal Medicine (J.W.Y.), University of Nevada Las Vegas School of Medicine; Department of Statistics and Actuarial Science (K.N.L., K.H.), Soongsil University; and Department of Biostatistics (J.-H.J., Y.-G.P.), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Do-Young Kwon
- From the Departments of Family Medicine (J.-H.P., C.-w.L., M.J.N., H.K., D.-H.K.) and Neurology (D.-Y.K.), Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea; Department of Internal Medicine (J.W.Y.), University of Nevada Las Vegas School of Medicine; Department of Statistics and Actuarial Science (K.N.L., K.H.), Soongsil University; and Department of Biostatistics (J.-H.J., Y.-G.P.), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Won Yoo
- From the Departments of Family Medicine (J.-H.P., C.-w.L., M.J.N., H.K., D.-H.K.) and Neurology (D.-Y.K.), Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea; Department of Internal Medicine (J.W.Y.), University of Nevada Las Vegas School of Medicine; Department of Statistics and Actuarial Science (K.N.L., K.H.), Soongsil University; and Department of Biostatistics (J.-H.J., Y.-G.P.), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyu Na Lee
- From the Departments of Family Medicine (J.-H.P., C.-w.L., M.J.N., H.K., D.-H.K.) and Neurology (D.-Y.K.), Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea; Department of Internal Medicine (J.W.Y.), University of Nevada Las Vegas School of Medicine; Department of Statistics and Actuarial Science (K.N.L., K.H.), Soongsil University; and Department of Biostatistics (J.-H.J., Y.-G.P.), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyungdo Han
- From the Departments of Family Medicine (J.-H.P., C.-w.L., M.J.N., H.K., D.-H.K.) and Neurology (D.-Y.K.), Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea; Department of Internal Medicine (J.W.Y.), University of Nevada Las Vegas School of Medicine; Department of Statistics and Actuarial Science (K.N.L., K.H.), Soongsil University; and Department of Biostatistics (J.-H.J., Y.-G.P.), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Jin-Hyung Jung
- From the Departments of Family Medicine (J.-H.P., C.-w.L., M.J.N., H.K., D.-H.K.) and Neurology (D.-Y.K.), Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea; Department of Internal Medicine (J.W.Y.), University of Nevada Las Vegas School of Medicine; Department of Statistics and Actuarial Science (K.N.L., K.H.), Soongsil University; and Department of Biostatistics (J.-H.J., Y.-G.P.), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yong-Gyu Park
- From the Departments of Family Medicine (J.-H.P., C.-w.L., M.J.N., H.K., D.-H.K.) and Neurology (D.-Y.K.), Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea; Department of Internal Medicine (J.W.Y.), University of Nevada Las Vegas School of Medicine; Department of Statistics and Actuarial Science (K.N.L., K.H.), Soongsil University; and Department of Biostatistics (J.-H.J., Y.-G.P.), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Do-Hoon Kim
- From the Departments of Family Medicine (J.-H.P., C.-w.L., M.J.N., H.K., D.-H.K.) and Neurology (D.-Y.K.), Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea; Department of Internal Medicine (J.W.Y.), University of Nevada Las Vegas School of Medicine; Department of Statistics and Actuarial Science (K.N.L., K.H.), Soongsil University; and Department of Biostatistics (J.-H.J., Y.-G.P.), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
18
|
Pedrini S, Chatterjee P, Hone E, Martins RN. High‐density lipoprotein‐related cholesterol metabolism in Alzheimer’s disease. J Neurochem 2020; 159:343-377. [DOI: 10.1111/jnc.15170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Steve Pedrini
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
| | - Pratishtha Chatterjee
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
- Department of Biomedical Sciences Faculty of Medicine, Health and Human Sciences Macquarie University Sydney NSW Australia
| | - Eugene Hone
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
| | - Ralph N. Martins
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
- Department of Biomedical Sciences Faculty of Medicine, Health and Human Sciences Macquarie University Sydney NSW Australia
- School of Psychiatry and Clinical Neurosciences University of Western Australia Nedlands WA Australia
| |
Collapse
|
19
|
Millwood IY, Bennett DA, Holmes MV, Boxall R, Guo Y, Bian Z, Yang L, Sansome S, Chen Y, Du H, Yu C, Hacker A, Reilly DF, Tan Y, Hill MR, Chen J, Peto R, Shen H, Collins R, Clarke R, Li L, Walters RG, Chen Z. Association of CETP Gene Variants With Risk for Vascular and Nonvascular Diseases Among Chinese Adults. JAMA Cardiol 2019; 3:34-43. [PMID: 29141072 PMCID: PMC5833522 DOI: 10.1001/jamacardio.2017.4177] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Question What is the association of genetic variants in the CETP gene that
lower cholesteryl ester transfer protein activity with risk for cardiovascular and other
diseases? Findings In this biobank study of 151 217 Chinese adults, CETP gene
variants were associated with higher levels of high-density lipoprotein cholesterol but
not with lower levels of low-density lipoprotein cholesterol and were not associated
with risk for cardiovascular disease. Meaning Increasing levels of high-density lipoprotein cholesterol by cholesteryl ester transfer
protein inhibition in the absence of lower levels of low-density lipoprotein cholesterol
may not confer significant benefits for cardiovascular disease. Importance Increasing levels of high-density lipoprotein (HDL) cholesterol through pharmacologic
inhibition of cholesteryl ester transfer protein (CETP) is a potentially important
strategy for prevention and treatment of cardiovascular disease (CVD). Objective To use genetic variants in the CETP gene to assess potential risks and
benefits of lifelong lower CETP activity on CVD and other outcomes. Design, Setting, and Participants This prospective biobank study included 151 217 individuals aged 30 to 79 years
who were enrolled from 5 urban and 5 rural areas of China from June 25, 2004, through
July 15, 2008. All participants had baseline genotype data, 17 854 of whom had
lipid measurements and 4657 of whom had lipoprotein particle measurements. Median
follow-up of 9.2 years (interquartile range, 8.2-10.1 years) was completed January 1,
2016, through linkage to health insurance records and death and disease registries. Exposures Five CETP variants, including an East Asian loss-of-function variant
(rs2303790), combined in a genetic score weighted to associations with HDL
cholesterol levels. Main Outcomes and Measures Baseline levels of lipids and lipoprotein particles, cardiovascular risk factors,
incidence of carotid plaque and predefined major vascular and nonvascular diseases, and
a phenome-wide range of diseases. Results Among the 151 217 individuals included in this study (58.4% women and 41.6% men),
the mean (SD) age was 52.3 (10.9) years. Overall, the mean (SD) low-density lipoprotein
(LDL) cholesterol level was 91 (27) mg/dL; HDL cholesterol level, 48 (12) mg/dL.
CETP variants were strongly associated with higher concentrations of
HDL cholesterol (eg, 6.1 [SE, 0.4] mg/dL per rs2303790-G
allele; P = 9.4 × 10−47)
but were not associated with lower LDL cholesterol levels. Within HDL particles,
cholesterol esters were increased and triglycerides reduced, whereas within very
low-density lipoprotein particles, cholesterol esters were reduced and triglycerides
increased. When scaled to 10-mg/dL higher levels of HDL cholesterol, the
CETP genetic score was not associated with occlusive CVD
(18 550 events; odds ratio [OR], 0.98; 95% CI, 0.91-1.06), major coronary events
(5767 events; OR, 1.08; 95% CI, 0.95-1.22), myocardial infarction (3118 events; OR,
1.14; 95% CI, 0.97-1.35), ischemic stroke (13 759 events; OR, 0.94; 95% CI,
0.86-1.02), intracerebral hemorrhage (6532 events; OR, 0.94; 95% CI, 0.83-1.06), or
other vascular diseases or carotid plaque. Similarly, rs2303790 was not
associated with any vascular diseases or plaque. No associations with nonvascular
diseases were found other than an increased risk for eye diseases with rs2303790 (4090 events; OR, 1.43; 95% CI, 1.13-1.80;
P = .003). Conclusions and Relevance CETP variants were associated with altered HDL metabolism but did not
lower LDL cholesterol levels and had no significant association with risk for CVD. These
results suggest that in the absence of reduced LDL cholesterol levels, increasing HDL
cholesterol levels by inhibition of CETP may not confer significant benefits for
CVD.
Collapse
Affiliation(s)
- Iona Y Millwood
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England.,Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England
| | - Derrick A Bennett
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England
| | - Michael V Holmes
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England.,Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital, Oxford, England
| | - Ruth Boxall
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England.,Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England
| | - Yu Guo
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China
| | - Zheng Bian
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China
| | - Ling Yang
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England.,Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England
| | - Sam Sansome
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England
| | - Yiping Chen
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England.,Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England
| | - Huaidong Du
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England.,Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England
| | - Canqing Yu
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China
| | - Alex Hacker
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England
| | | | - Yunlong Tan
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China
| | - Michael R Hill
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England
| | - Junshi Chen
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China
| | - Richard Peto
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, Nanjing Medical University School of Public Health, Nanjing, China
| | - Rory Collins
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England
| | - Robert Clarke
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England
| | - Liming Li
- Department of Epidemiology and Biostatistics, Peking University Health Science Centre, Peking University, Beijing, China
| | - Robin G Walters
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England
| | | |
Collapse
|
20
|
Kinoshita M, Yokote K, Arai H, Iida M, Ishigaki Y, Ishibashi S, Umemoto S, Egusa G, Ohmura H, Okamura T, Kihara S, Koba S, Saito I, Shoji T, Daida H, Tsukamoto K, Deguchi J, Dohi S, Dobashi K, Hamaguchi H, Hara M, Hiro T, Biro S, Fujioka Y, Maruyama C, Miyamoto Y, Murakami Y, Yokode M, Yoshida H, Rakugi H, Wakatsuki A, Yamashita S. Japan Atherosclerosis Society (JAS) Guidelines for Prevention of Atherosclerotic Cardiovascular Diseases 2017. J Atheroscler Thromb 2018; 25:846-984. [PMID: 30135334 PMCID: PMC6143773 DOI: 10.5551/jat.gl2017] [Citation(s) in RCA: 511] [Impact Index Per Article: 85.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/11/2018] [Indexed: 11/30/2022] Open
Affiliation(s)
| | - Koutaro Yokote
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hidenori Arai
- National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Mami Iida
- Department of Internal Medicine and Cardiology, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Yasushi Ishigaki
- Division of Diabetes and Metabolism, Department of Internal Medicine, Iwate Medical University, Iwate, Japan
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Seiji Umemoto
- Center for Integrated Medical Research, Hiroshima University Hospital, Hiroshima, Japan
| | | | - Hirotoshi Ohmura
- Department of Cardiovascular Medicine, Juntendo University, Tokyo, Japan
| | - Tomonori Okamura
- Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Shinji Kihara
- Biomedical Informatics, Osaka University, Osaka, Japan
| | - Shinji Koba
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Isao Saito
- Department of Community Health Systems Nursing, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Tetsuo Shoji
- Department of Vascular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hiroyuki Daida
- Department of Cardiovascular Medicine, Juntendo University, Tokyo, Japan
| | - Kazuhisa Tsukamoto
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Juno Deguchi
- Department of Vascular Surgery, Saitama Medical Center, Saitama, Japan
| | - Seitaro Dohi
- Chief Health Management Department, Mitsui Chemicals Inc., Tokyo, Japan
| | - Kazushige Dobashi
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | | | - Masumi Hara
- Department of Internal Medicine, Mizonokuchi Hospital, Teikyo University School of Medicine, Kanagawa, Japan
| | - Takafumi Hiro
- Division of Cardiology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | | | - Yoshio Fujioka
- Faculty of Nutrition, Division of Clinical Nutrition, Kobe Gakuin University, Hyogo, Japan
| | - Chizuko Maruyama
- Department of Food and Nutrition, Faculty of Human Sciences and Design, Japan Women's University, Tokyo, Japan
| | - Yoshihiro Miyamoto
- Department of Preventive Cardiology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | | | - Masayuki Yokode
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Yoshida
- Department of Laboratory Medicine, Jikei University Kashiwa Hospital, Chiba, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akihiko Wakatsuki
- Department of Obstetrics and Gynecology, Aichi Medical University, Aichi, Japan
| | - Shizuya Yamashita
- Department of Community Medicine, Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Rinku General Medical Center, Osaka, Japan
| | | |
Collapse
|
21
|
Shrestha S, Wu BJ, Guiney L, Barter PJ, Rye KA. Cholesteryl ester transfer protein and its inhibitors. J Lipid Res 2018; 59:772-783. [PMID: 29487091 PMCID: PMC5928430 DOI: 10.1194/jlr.r082735] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/10/2018] [Indexed: 12/22/2022] Open
Abstract
Most of the cholesterol in plasma is in an esterified form that is generated in potentially cardioprotective HDLs. Cholesteryl ester transfer protein (CETP) mediates bidirectional transfers of cholesteryl esters (CEs) and triglycerides (TGs) between plasma lipoproteins. Because CE originates in HDLs and TG enters the plasma as a component of VLDLs, activity of CETP results in a net mass transfer of CE from HDLs to VLDLs and LDLs, and of TG from VLDLs to LDLs and HDLs. As inhibition of CETP activity increases the concentration of HDL-cholesterol and decreases the concentration of VLDL- and LDL-cholesterol, it has the potential to reduce atherosclerotic CVD. This has led to the development of anti-CETP neutralizing monoclonal antibodies, vaccines, and antisense oligonucleotides. Small molecule inhibitors of CETP have also been developed and four of them have been studied in large scale cardiovascular clinical outcome trials. This review describes the structure of CETP and its mechanism of action. Details of its regulation and nonlipid transporting functions are discussed, and the results of the large scale clinical outcome trials of small molecule CETP inhibitors are summarized.
Collapse
Affiliation(s)
- Sudichhya Shrestha
- School of Medical Sciences, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Ben J Wu
- School of Medical Sciences, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Liam Guiney
- Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Philip J Barter
- School of Medical Sciences, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Kerry-Anne Rye
- School of Medical Sciences, University of New South Wales Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
22
|
Sasaki M, Harada S, Kawasaki Y, Watanabe M, Ito H, Tanaka H, Takeuchi A, Tsubota K, Takebayashi T, Nishiwaki Y, Kawasaki R. Gender-specific association of early age-related macular degeneration with systemic and genetic factors in a Japanese population. Sci Rep 2018; 8:785. [PMID: 29335418 PMCID: PMC5768785 DOI: 10.1038/s41598-017-18487-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/12/2017] [Indexed: 01/12/2023] Open
Abstract
The Tsuruoka Metabolomics Cohort Study included subjects aged 35–74 years from participants in annual health check-up programs in Tsuruoka, Japan. The gender-specific associations of early age-related macular degeneration (AMD) with systemic and genetic factors was assessed cross-sectionally. Of these, 3,988 subjects had fundus photographs of sufficient quality, and early AMD was present in 12.3% and 10.3% of men and women, respectively. In men, higher levels of high-density lipoprotein cholesterol and lower levels of triglycerides were associated with increased odds of having early AMD after adjusting for potential risk factors (for each 1 mmol/L increase, odds ratio [OR]: 1.61 and 0.78, 95% confidence interval [CI]: 1.17–2.23 and 0.64–0.96, respectively). In women, higher levels of total cholesterol and low-density lipoprotein cholesterol were associated with increased risk of having early AMD (OR: 1.21 and 1.26, 95% CI: 1.01–1.44 and 1.03–1.53, respectively). Sub-analysis demonstrated that women with ARMS2 A69S polymorphisms had a stronger risk for early AMD (OR: 3.25, 95% CI: 2.10–5.04) than men (OR: 1.65, 95% CI: 1.02–2.69). Differential associations of early AMD with both systemic and genetic factors by sex were demonstrated in a Japanese cohort, which suggests that disease process of early AMD could be different by sex.
Collapse
Affiliation(s)
- Mariko Sasaki
- Keio University, Department of Ophthalmology, 160-0016, Tokyo, Japan.,Tachikawa Hospital, 190-8531, Tokyo, Japan.,National Institute of Sensory Organs, National Tokyo Medical Center, 152-8902, Tokyo, Japan
| | - Sei Harada
- Department of Preventive Medicine and Public Health, Keio University, 160-8582, Tokyo, Japan
| | - Yumiko Kawasaki
- Yamagata University Graduate School of Medical Science, Department of Public Health, 990-2331, Yamagata, Japan
| | - Miki Watanabe
- Nagoya City University Graduate School of Medical Sciences, Department of Public Health, 467-8601, Aichi, Japan
| | - Hidemi Ito
- Aichi Cancer Research Institute, Division of Molecular & Clinical Epidemiology, 464-8681, Aichi, Japan
| | - Hideo Tanaka
- Kishiwada Public Health Center, 596-0076, Osaka, Japan
| | - Ayano Takeuchi
- Department of Preventive Medicine and Public Health, Keio University, 160-8582, Tokyo, Japan
| | - Kazuo Tsubota
- Keio University, Department of Ophthalmology, 160-0016, Tokyo, Japan
| | - Toru Takebayashi
- Department of Preventive Medicine and Public Health, Keio University, 160-8582, Tokyo, Japan
| | - Yuji Nishiwaki
- Toho University, Department of Environmental and Occupational Health, 143-8540, Tokyo, Japan
| | - Ryo Kawasaki
- Yamagata University Graduate School of Medical Science, Department of Public Health, 990-2331, Yamagata, Japan.
| |
Collapse
|
23
|
Vitali C, Khetarpal SA, Rader DJ. HDL Cholesterol Metabolism and the Risk of CHD: New Insights from Human Genetics. Curr Cardiol Rep 2017; 19:132. [PMID: 29103089 DOI: 10.1007/s11886-017-0940-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW Elevated high-density lipoprotein cholesterol levels in the blood (HDL-C) represent one of the strongest epidemiological surrogates for protection against coronary heart disease (CHD), but recent human genetic and pharmacological intervention studies have raised controversy about the causality of this relationship. Here, we review recent discoveries from human genome studies using new analytic tools as well as relevant animal studies that have both addressed, and in some cases, fueled this controversy. RECENT FINDINGS Methodologic developments in genotyping and sequencing, such as genome-wide association studies (GWAS), exome sequencing, and exome array genotyping, have been applied to the study of HDL-C and risk of CHD in large, multi-ethnic populations. Some of these efforts focused on population-wide variation in common variants have uncovered new polymorphisms at novel loci associated with HDL-C and, in some cases, CHD risk. Other efforts have discovered loss-of-function variants for the first time in genes previously implicated in HDL metabolism through common variant studies or animal models. These studies have allowed the genetic relationship between these pathways, HDL-C and CHD to be explored in humans for the first time through analysis tools such as Mendelian randomization. We explore these discoveries for selected key HDL-C genes CETP, LCAT, LIPG, SCARB1, and novel loci implicated from GWAS including GALNT2, KLF14, and TTC39B. Recent human genetics findings have identified new nodes regulating HDL metabolism while reshaping our current understanding of known candidate genes to HDL and CHD risk through the study of critical variants across model systems. Despite their effect on HDL-C, variants in many of the reviewed genes were found to lack any association with CHD. These data collectively indicate that HDL-C concentration, which represents a static picture of a very dynamic and heterogeneous metabolic milieu, is unlikely to be itself causally protective against CHD. In this context, human genetics represent an extremely valuable tool to further explore the biological mechanisms regulating HDL metabolism and investigate what role, if any, HDL plays in the pathogenesis of CHD.
Collapse
Affiliation(s)
- Cecilia Vitali
- Perelman School of Medicine at the University of Pennsylvania, 11-162 TRC, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Sumeet A Khetarpal
- Perelman School of Medicine at the University of Pennsylvania, 11-162 TRC, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Daniel J Rader
- Perelman School of Medicine at the University of Pennsylvania, 11-162 TRC, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA. .,Departments of Genetics and Medicine, Cardiovascular Institute, and Institute for Translational Medicine and Therapeutics, Perelman School of Medicine at the University of Pennsylvania, 11-125 TRC, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| |
Collapse
|
24
|
Cheung CMG, Gan A, Fan Q, Chee ML, Apte RS, Khor CC, Yeo I, Mathur R, Cheng CY, Wong TY, Tai ES. Plasma lipoprotein subfraction concentrations are associated with lipid metabolism and age-related macular degeneration. J Lipid Res 2017; 58:1785-1796. [PMID: 28698208 PMCID: PMC5580892 DOI: 10.1194/jlr.m073684] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 07/06/2017] [Indexed: 01/09/2023] Open
Abstract
Disturbance in lipid metabolism has been suggested as a major pathogenic factor for age-related macular degeneration (AMD). Conventional lipid measures have been inconsistently associated with AMD. Other factors that can alter lipid metabolism include lipoprotein phenotype and genetic mutations. We performed a case-control study to examine the association between lipoprotein profile and neovascular AMD (nAMD) and whether the cholesterylester transfer protein (CETP) D442G mutation modulates these associations. Patients with nAMD had significantly higher concentrations of HDL and IDL compared with controls. The increase in HDL particles in nAMD patients was driven by an excess of medium-sized particles. Concurrently, patients with nAMD also had lower Apo A-1, lower VLDL and chylomicron lipoprotein. Many of these associations showed a dose-dependent association between controls, early AMD cases, and nAMD cases. Adjustment for the presence of the D442G mutation at the CETP locus did not significantly alter the increased AMD risk associated with HDL particle concentration. AMD is associated with variation in many lipoprotein subclasses, including increased HDL and IDL particles and decreased Apo A-1, VLDL, and chylomicron particles. These data suggest widespread systemic disturbance in lipid metabolism in the pathogenesis of AMD, including possible alterations in lipoprotein carrier capacity.
Collapse
Affiliation(s)
- Chui Ming Gemmy Cheung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Ophthalmology and Visual Sciences Program, Duke-NUS Medical School, National University of Singapore, Singapore.
| | - Alfred Gan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Qiao Fan
- Centre for Quantitative Medicine, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Miao Ling Chee
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Rajendra S Apte
- Ophthalmology and Visual Sciences, Developmental Biology and Medicine, Washington University School of Medicine, St. Louis, MO
| | | | - Ian Yeo
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Program, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Ranjana Mathur
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Program, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Program, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Ophthalmology and Visual Sciences Program, Duke-NUS Medical School, National University of Singapore, Singapore
| | - E Shyong Tai
- Department of Medicine, Cardiovascular and Metabolic Disorders Programme, National University of Singapore, Singapore
| |
Collapse
|
25
|
Nomura A, Won HH, Khera AV, Takeuchi F, Ito K, McCarthy S, Emdin CA, Klarin D, Natarajan P, Zekavat SM, Gupta N, Peloso GM, Borecki IB, Teslovich TM, Asselta R, Duga S, Merlini PA, Correa A, Kessler T, Wilson JG, Bown MJ, Hall AS, Braund PS, Carey DJ, Murray MF, Kirchner HL, Leader JB, Lavage DR, Manus JN, Hartze DN, Samani NJ, Schunkert H, Marrugat J, Elosua R, McPherson R, Farrall M, Watkins H, Juang JMJ, Hsiung CA, Lin SY, Wang JS, Tada H, Kawashiri MA, Inazu A, Yamagishi M, Katsuya T, Nakashima E, Nakatochi M, Yamamoto K, Yokota M, Momozawa Y, Rotter JI, Lander ES, Rader DJ, Danesh J, Ardissino D, Gabriel S, Willer CJ, Abecasis GR, Saleheen D, Kubo M, Kato N, Ida Chen YD, Dewey FE, Kathiresan S. Protein-Truncating Variants at the Cholesteryl Ester Transfer Protein Gene and Risk for Coronary Heart Disease. Circ Res 2017; 121:81-88. [PMID: 28506971 PMCID: PMC5523940 DOI: 10.1161/circresaha.117.311145] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 04/28/2017] [Accepted: 05/12/2017] [Indexed: 12/30/2022]
Abstract
RATIONALE Therapies that inhibit CETP (cholesteryl ester transfer protein) have failed to demonstrate a reduction in risk for coronary heart disease (CHD). Human DNA sequence variants that truncate the CETP gene may provide insight into the efficacy of CETP inhibition. OBJECTIVE To test whether protein-truncating variants (PTVs) at the CETP gene were associated with plasma lipid levels and CHD. METHODS AND RESULTS We sequenced the exons of the CETP gene in 58 469 participants from 12 case-control studies (18 817 CHD cases, 39 652 CHD-free controls). We defined PTV as those that lead to a premature stop, disrupt canonical splice sites, or lead to insertions/deletions that shift frame. We also genotyped 1 Japanese-specific PTV in 27561 participants from 3 case-control studies (14 286 CHD cases, 13 275 CHD-free controls). We tested association of CETP PTV carrier status with both plasma lipids and CHD. Among 58 469 participants with CETP gene-sequencing data available, average age was 51.5 years and 43% were women; 1 in 975 participants carried a PTV at the CETP gene. Compared with noncarriers, carriers of PTV at CETP had higher high-density lipoprotein cholesterol (effect size, 22.6 mg/dL; 95% confidence interval, 18-27; P<1.0×10-4), lower low-density lipoprotein cholesterol (-12.2 mg/dL; 95% confidence interval, -23 to -0.98; P=0.033), and lower triglycerides (-6.3%; 95% confidence interval, -12 to -0.22; P=0.043). CETP PTV carrier status was associated with reduced risk for CHD (summary odds ratio, 0.70; 95% confidence interval, 0.54-0.90; P=5.1×10-3). CONCLUSIONS Compared with noncarriers, carriers of PTV at CETP displayed higher high-density lipoprotein cholesterol, lower low-density lipoprotein cholesterol, lower triglycerides, and lower risk for CHD.
Collapse
|
26
|
Yang X, Sethi A, Yanek LR, Knapper C, Nordestgaard BG, Tybjærg-Hansen A, Becker DM, Mathias RA, Remaley AT, Becker LC. SCARB1 Gene Variants Are Associated With the Phenotype of Combined High High-Density Lipoprotein Cholesterol and High Lipoprotein (a). ACTA ACUST UNITED AC 2016; 9:408-418. [PMID: 27651445 DOI: 10.1161/circgenetics.116.001402] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 08/20/2016] [Indexed: 02/03/2023]
Abstract
BACKGROUND SR-B1 (scavenger receptor class B type 1), encoded by the gene SCARB1, is a lipoprotein receptor that binds both high-density lipoprotein (HDL) and low-density lipoprotein. We reported that SR-B1 is also a receptor for lipoprotein (a) (Lp(a)), mediating cellular uptake of Lp(a) in vitro and promoting clearance of Lp(a) in vivo. Although genetic variants in SCARB1 are associated with variations in HDL level, no SCARB1 variants affecting Lp(a) have been reported. METHODS AND RESULTS In an index subject with high levels of HDL cholesterol and Lp(a), SCARB1 was sequenced and demonstrated a missense mutation resulting in an S129L substitution in exon 3. To follow up, 2 cohorts (GeneSTAR, the family-based Genetic Study of Atherosclerosis Risk [n=543], and CCHS, the population-based Copenhagen City Heart Study [n=5835]) were screened for combined HDL cholesterol and Lp(a) elevations. Subjects with the extreme phenotype (HDL >80 mg/dL and Lp(a) >100 nmol/L in GeneSTAR, n=8, and >100 mg/dL in CCHS, n=9) underwent sequencing of SCARB1 exons; 15 of 18 from the combined population demonstrated genetic variants, including rare or uncommon missense or splice site mutations in 9 and homozygous synonymous variants in 6. Functional studies with 4 of the SCARB1 variants (c.386C>T, c.631-14T>G, c.4G>A, and c.631-53mC>T & c.726+55mCG>CA) showed decreased receptor function in vitro. CONCLUSIONS Human SCARB1 gene variants are associated with a new lipid phenotype, characterized by high levels of both HDL cholesterol and Lp(a). SCARB1 exonic variants often result in diminished function of translated SR-B1 via reduced binding/intracellular transport of Lp(a).
Collapse
Affiliation(s)
- Xiaoping Yang
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Amar Sethi
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Lisa R Yanek
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Cathy Knapper
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Børge G Nordestgaard
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Anne Tybjærg-Hansen
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Diane M Becker
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Rasika A Mathias
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Alan T Remaley
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Lewis C Becker
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.).
| |
Collapse
|
27
|
Abstract
The cholesteryl ester transfer protein (CETP) is a plasma protein that plays an important role in the transfer of lipids between plasma lipoproteins. The CETP inhibitors have been widely studied as a pharmacologic therapy to target plasma cholesterol in order to reduce the risk of atherosclerotic cardiovascular disease . Using CETP inhibitors as cholesterol modifiers was based on the genetic research that found correlations between CETP activity and cholesterol levels. Although CETP inhibitors are successful at altering targeted cholesterol markers, recent phase 3 outcome trials have shown limited benefit on cardiovascular outcomes when combined with the current standard of care. We discuss the science of CETP inhibition, compare the CETP inhibitors developed (torcetrapib, evacetrapib, dalcetrapib, and anacetrapib), the findings from the CETP inhibitor trials, and the future outlook for CETP inhibitors in cholesterol modification.
Collapse
Affiliation(s)
- Julian Hardy McLain
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Andrew Jacob Alsterda
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Rohit R. Arora
- Captain James A. Lovell Federal Health Care Center, North Chicago, IL, USA
| |
Collapse
|
28
|
Bustami J, Sukiasyan A, Kupcinskas J, Skieceviciene J, Iakoubov L, Szwed M, Kleinle C, Schumann RR, Puzianowska-Kuznicka M, Hamann L. Cholesteryl ester transfer protein (CETP) I405V polymorphism and cardiovascular disease in eastern European Caucasians - a cross-sectional study. BMC Geriatr 2016; 16:144. [PMID: 27439317 PMCID: PMC4955243 DOI: 10.1186/s12877-016-0318-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/14/2016] [Indexed: 11/13/2022] Open
Abstract
Background The cholesteryl ester transfer protein (CETP) polymorphism I405V has been suggested to be involved in longevity and susceptibility to cardiovascular diseases. An enhanced reverse cholesterol transport due to enhanced HDL levels has been hypothesized to be the underlying mechanism. However, clinical trials with HDL-enhancing drugs failed to show beneficial effects. Consequently, it has been postulated that genetic variations enhancing HDL levels are cardioprotective only if they also decrease LDL levels. Methods A cross-sectional study was conducted to genotype 1028 healthy blood donors and 1517 clinically well characterized elderly for CETP I405V. Results We could not find any association of this polymorphism with age for both, males or females, in any of these cohorts (P = 0.71 and P = 0.57, respectively, for males and P = 0.55 and P = 0.88, respectively, for females). In addition, no association with cardiovascular diseases could be observed in the elderly cohort (males OR = 1.12 and females OR = 0.88). In the same cohort, the CETP V405V genotype was associated with significantly enhanced HDL levels (P = 0.03), mostly owing to the female sex (P = 0.46 for males, P = 0.02 for females), whereas LDL and triglyceride levels were unchanged (P = 0.62 and P = 0.18, respectively). Conclusion Our data support the recent hypothesis that variations enhancing HDL levels without affecting LDL levels are not associated with the risk for cardiovascular diseases.
Collapse
Affiliation(s)
- Jasmin Bustami
- Institute of Microbiology and Hygiene, Charité University Medical Center Berlin, Rahel-Hirsch-Weg 3, 10117, Berlin, Germany
| | - Anna Sukiasyan
- Institute of Microbiology and Hygiene, Charité University Medical Center Berlin, Rahel-Hirsch-Weg 3, 10117, Berlin, Germany.,Research Institute of Epidemiology, MOH, Yerevan, Armenia
| | - Juozas Kupcinskas
- Department of Gastro-enterology, Lithuanian University of Health Sciences, Kaunas, Lithuania.,Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Jurgita Skieceviciene
- Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | - Malgorzata Szwed
- Department of Human Epigenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Christoph Kleinle
- Institute of Microbiology and Hygiene, Charité University Medical Center Berlin, Rahel-Hirsch-Weg 3, 10117, Berlin, Germany
| | - Ralf R Schumann
- Institute of Microbiology and Hygiene, Charité University Medical Center Berlin, Rahel-Hirsch-Weg 3, 10117, Berlin, Germany
| | - Monika Puzianowska-Kuznicka
- Department of Human Epigenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Department of Geriatrics and Gerontology, Medical Centre of Postgraduate Education, Warsaw, Poland
| | - Lutz Hamann
- Institute of Microbiology and Hygiene, Charité University Medical Center Berlin, Rahel-Hirsch-Weg 3, 10117, Berlin, Germany.
| |
Collapse
|
29
|
Quintão ECR. The controversy over the use of cholesteryl ester transfer protein inhibitors: is there some light at the end of the tunnel? Eur J Clin Invest 2016; 46:581-9. [PMID: 26992444 DOI: 10.1111/eci.12626] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/16/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND According to epidemiological studies, there is no clear relationship between the plasma cholesteryl ester transfer protein (CETP) concentration and the development of atherosclerosis in human populations. Although some studies suggest that increased CETP activity relates to undesirable profiles of plasma lipoproteins, promoting an anti-atherogenic plasma lipoprotein profile by drugs that inhibit CETP has not succeeded in preventing atherosclerosis in humans. MATERIALS AND METHODS This review describes 28 investigations in human populations dealing with plasma CETP, 11 in mice that express human CETP and seven in animals (six in rabbits and one in mice) in which plasma CETP activity was inhibited by drugs. RESULTS Present review shows that models in mice expressing human CETP are not illuminating because they report increase as well reduction of atherosclerosis. However, investigations in rabbits and mice that develop severe hypercholesterolaemia clearly indicate that impairment of the plasma CETP activity elicits protection against the development of atherosclerosis; in all of these experiments are attained substantial reductions of the atherogenic lipoproteins, namely, plasma apoB containing lipoproteins. CONCLUSION These models are strong indicators that the benefit in preventing atherosclerosis should be earned in cases of hyperlipidemia by CETP inhibitors.
Collapse
Affiliation(s)
- Eder C R Quintão
- Internal Medicine, University of Sao Paulo Medical School, Sao Paulo, Brazil
| |
Collapse
|
30
|
Filippatos TD, Klouras E, Barkas F, Elisaf M. Cholesteryl ester transfer protein inhibitors: challenges and perspectives. Expert Rev Cardiovasc Ther 2016; 14:953-62. [DOI: 10.1080/14779072.2016.1189327] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
31
|
Wong CW, Yanagi Y, Lee WK, Ogura Y, Yeo I, Wong TY, Cheung CMG. Age-related macular degeneration and polypoidal choroidal vasculopathy in Asians. Prog Retin Eye Res 2016; 53:107-139. [PMID: 27094371 DOI: 10.1016/j.preteyeres.2016.04.002] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Revised: 03/31/2016] [Accepted: 04/11/2016] [Indexed: 12/12/2022]
Abstract
Age-related macular degeneration (AMD) is the leading cause of irreversible blindness in elderly people globally. It is estimated that there will be more Asians with AMD than the rest of the world combined by 2050. In Asian populations, polypoidal choroidal vasculopathy (PCV) is a common subtype of exudative AMD, while choroidal neovascularization secondary to AMD (CNV-AMD) is the typical subtype in Western populations. The two subtypes share many common clinical features and risk factors, but also have different epidemiological and clinical characteristics, natural history and treatment outcomes that point to distinct pathophysiological processes. Recent research in the fields of genetics, proteomics and imaging has provided further clarification of differences between PCV and CNV-AMD. Importantly, these differences have manifested as disparity in response to intravitreal injections of anti-vascular endothelial growth factor (anti-VEGF) treatment between PCV and CNV-AMD, emphasizing the need for accurate diagnosis of PCV and in distinguishing PCV from CNV-AMD, particularly in Asian patients. Current clinical trials of intravitreal anti-VEGF therapy and photodynamic therapy will provide clearer perspectives of evidence-based management of PCV and may lead to paradigm shifts in therapeutic strategies away from those currently employed in the treatment of CNV-AMD. Further research is needed to clarify the relative contribution of specific pathways in inflammation, complement activation, extracellular matrix dysregulation, lipid metabolism and angiogenesis to the pathogenesis of PCV. Findings from this research, together with improved diagnostic technology and new therapeutics, will facilitate more optimal management of Asian AMD.
Collapse
Affiliation(s)
- Chee Wai Wong
- Singapore National Eye Centre, Singapore; Singapore Eye Research Institute, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Yasuo Yanagi
- Singapore National Eye Centre, Singapore; Singapore Eye Research Institute, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Won-Ki Lee
- Department of Ophthalmology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Yuichiro Ogura
- Department of Ophthalmology, Nagoya City University, Nagoya, Japan
| | - Ian Yeo
- Singapore National Eye Centre, Singapore; Singapore Eye Research Institute, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Tien Yin Wong
- Singapore National Eye Centre, Singapore; Singapore Eye Research Institute, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore.
| | - Chui Ming Gemmy Cheung
- Singapore National Eye Centre, Singapore; Singapore Eye Research Institute, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| |
Collapse
|
32
|
Kurano M, Tsukamoto K, Kamitsuji S, Kamatani N, Hara M, Ishikawa T, Kim BJ, Moon S, Jin Kim Y, Teramoto T. Genome-wide association study of serum lipids confirms previously reported associations as well as new associations of common SNPs within PCSK7 gene with triglyceride. J Hum Genet 2016; 61:427-33. [DOI: 10.1038/jhg.2015.170] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/23/2015] [Accepted: 12/13/2015] [Indexed: 12/31/2022]
|
33
|
Takahashi Y, Ito Y, Wada N, Nagasaka A, Fujikawa M, Sakurai T, Shrestha R, Hui SP, Chiba H. Development of homogeneous assay for simultaneous measurement of apoE-deficient, apoE-containing, and total HDL-cholesterol. Clin Chim Acta 2016; 454:135-42. [PMID: 26776838 DOI: 10.1016/j.cca.2016.01.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 01/12/2016] [Accepted: 01/12/2016] [Indexed: 11/25/2022]
Abstract
BACKGROUND Pathophysiological role for high-density lipoprotein (HDL) subclasses remains to be elucidated. Homogeneous assay for simultaneous measurements of apoE-deficient HDL-cholesterol (HDL-C), apoE-containing HDL-C, and total HDL-C is desired, because apoE plays important roles in lipid metabolism. METHODS The proposed assay consists of a primary reaction to remove non-HDL-C, a secondary reaction to measure apoE-deficient HDL-C, and a tertiary reaction to measure apoE-containing HDL-C. The assay is completed within 10 min. For control study, 13% polyethylene glycol precipitation assay and phosphotungstate-dextran sulfate-magnesium precipitation assay were carried out. RESULTS Good correlations between the control assays and the proposed assay was obtained in serum samples from patients without liver disease (n=33): r=0.987, 0.957, and 0.991 for apoE-deficient, apoE-containing, and total HDL-C, respectively. ApoE-containing HDL-C by the proposed method in healthy individuals (n=12) and patients with hyper-HDL-cholesterolemia (n=5) were 0.11±0.03 and 0.26±0.05 mmol/l (4.1±1.3 and 10.1±2.0 mg/dl), respectively. ApoE-containing HDL-C increased rapidly at >2.59 mmol/l (100 mg/dl) of total HDL-C, suggesting a unique regulating mechanism of apoE-containing HDL-C. CONCLUSIONS The established homogeneous assay might be useful for clinical and epidemiological studies on apoE-deficient and apoE-containing HDL subclasses.
Collapse
Affiliation(s)
- Yuji Takahashi
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan; Department of Clinical Laboratory, Sapporo City General Hospital, Sapporo, Japan
| | - Yasuki Ito
- Reagent R & D Department, Denka Seiken Co., Ltd., Niigata, Japan
| | - Norio Wada
- Department of Diabetes and Endocrinology, Sapporo City General Hospital, Sapporo, Japan
| | - Atsushi Nagasaka
- Department of Infectious Diseases, Sapporo City General Hospital, Sapporo, Japan
| | - Masato Fujikawa
- Department of Clinical Laboratory, Sapporo City General Hospital, Sapporo, Japan
| | | | - Rojeet Shrestha
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Shu-Ping Hui
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Hitoshi Chiba
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
34
|
Dysfunctional High-Density Lipoprotein: An Innovative Target for Proteomics and Lipidomics. CHOLESTEROL 2015; 2015:296417. [PMID: 26634153 PMCID: PMC4655037 DOI: 10.1155/2015/296417] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 10/12/2015] [Accepted: 10/12/2015] [Indexed: 02/02/2023]
Abstract
High-Density Lipoprotein-Cholesterol (HDL-C) is regarded as an important protective factor against cardiovascular disease, with abundant evidence of an inverse relationship between its serum levels and risk of cardiovascular disease, as well as various antiatherogenic, antioxidant, and anti-inflammatory properties. Nevertheless, observations of hereditary syndromes featuring scant HDL-C concentration in absence of premature atherosclerotic disease suggest HDL-C levels may not be the best predictor of cardiovascular disease. Indeed, the beneficial effects of HDL may not depend solely on their concentration, but also on their quality. Distinct subfractions of this lipoprotein appear to be constituted by specific protein-lipid conglomerates necessary for different physiologic and pathophysiologic functions. However, in a chronic inflammatory microenvironment, diverse components of the HDL proteome and lipid core suffer alterations, which propel a shift towards a dysfunctional state, where HDL-C becomes proatherogenic, prooxidant, and proinflammatory. This heterogeneity highlights the need for further specialized molecular studies in this aspect, in order to achieve a better understanding of this dysfunctional state; with an emphasis on the potential role for proteomics and lipidomics as valuable methods in the search of novel therapeutic approaches for cardiovascular disease.
Collapse
|
35
|
Kathiresan S. Developing medicines that mimic the natural successes of the human genome: lessons from NPC1L1, HMGCR, PCSK9, APOC3, and CETP. J Am Coll Cardiol 2015; 65:1562-6. [PMID: 25881938 DOI: 10.1016/j.jacc.2015.02.049] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 02/24/2015] [Indexed: 11/18/2022]
Affiliation(s)
- Sekar Kathiresan
- Center for Human Genetic Research and Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts; Department of Medicine, Harvard Medical School, Boston, Massachusetts; and the Broad Institute of MIT and Harvard, Cambridge, Massachusetts.
| |
Collapse
|
36
|
Yokoyama S, Okumura-Noji K, Lu R. Prevention of fatal hepatic complication in schistosomiasis by inhibition of CETP. J Biomed Res 2015; 29:176-88. [PMID: 26060442 PMCID: PMC4449486 DOI: 10.7555/jbr.29.20150005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 01/26/2015] [Indexed: 11/04/2022] Open
Abstract
Schistosoma japonicum, once endemic all the East Asia, remains as a serious public health problem in certain regions. Ectopic egg embryonation in the liver causes granulomatosis and eventually fatal cirrhosis, so that prevention of this process is one of the keys to reduce its mortality. The embryonation requires cholesteryl ester from HDL of the host blood for egg yolk formation, and this reaction is impaired from the abnormal large HDL in genetic cholesteryl ester transfer protein (CETP) deficiency. When CETP was expressed in mice that otherwise lack this protein, granulomatosis of the liver was shown increased compared to the wild type upon infection of Schistosoma japonicum. The CETP deficiencies accumulated exclusively in East Asia, from Indochina to Siberia, so that Shistosomiasis can be a screening factor for this accumulation. CD36 related protein (CD36RP) was identified as a protein for this reaction, cloned from the cDNA library of Schistosoma japonicum with 1880-bp encoding 506 amino acids. The antibody against the extracellular loop of CD36RP inhibited cholesteryl ester uptake from HDL and suppressed egg embryonation in culture. Therefore, inhibition of CETP is a potential approach to prevent liver granulomatosis and thereby fatal liver cirrhosis in the infection of Schistosoma japonicum.
Collapse
Affiliation(s)
- Shinji Yokoyama
- Nutritional Health Science Research Center, Chubu University, Kasugai 487-8501, Japan.
| | - Kuniko Okumura-Noji
- Nutritional Health Science Research Center, Chubu University, Kasugai 487-8501, Japan.
| | - Rui Lu
- Nutritional Health Science Research Center, Chubu University, Kasugai 487-8501, Japan.
| |
Collapse
|
37
|
Yokoyama S. Unique features of high-density lipoproteins in the Japanese: in population and in genetic factors. Nutrients 2015; 7:2359-81. [PMID: 25849946 PMCID: PMC4425149 DOI: 10.3390/nu7042359] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/09/2015] [Accepted: 02/25/2015] [Indexed: 11/16/2022] Open
Abstract
Despite its gradual increase in the past several decades, the prevalence of atherosclerotic vascular disease is low in Japan. This is largely attributed to difference in lifestyle, especially food and dietary habits, and it may be reflected in certain clinical parameters. Plasma high-density lipoprotein (HDL) levels, a strong counter risk for atherosclerosis, are indeed high among the Japanese. Accordingly, lower HDL seems to contribute more to the development of coronary heart disease (CHD) than an increase in non-HDL lipoproteins at a population level in Japan. Interestingly, average HDL levels in Japan have increased further in the past two decades, and are markedly higher than in Western populations. The reasons and consequences for public health of this increase are still unknown. Simulation for the efficacy of raising HDL cholesterol predicts a decrease in CHD of 70% in Japan, greater than the extent by reducing low-density lipoprotein cholesterol predicted by simulation or achieved in a statin trial. On the other hand, a substantial portion of hyperalphalipoproteinemic population in Japan is accounted for by genetic deficiency of cholesteryl ester transfer protein (CETP), which is also commonly unique in East Asian populations. It is still controversial whether CETP mutations are antiatherogenic. Hepatic Schistosomiasis is proposed as a potential screening factor for historic accumulation of CETP deficiency in East Asia.
Collapse
Affiliation(s)
- Shinji Yokoyama
- Nutritional Health Science Research Centre and Bioscience and Biotechnology, Chubu University, Kasugai 487-8501, Japan.
| |
Collapse
|
38
|
Brunham LR, Hayden MR. Human genetics of HDL: Insight into particle metabolism and function. Prog Lipid Res 2015; 58:14-25. [DOI: 10.1016/j.plipres.2015.01.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/22/2014] [Accepted: 01/07/2015] [Indexed: 10/24/2022]
|
39
|
Kaman D, İlhan N, İlhan N, Akbulut M. TaqIB and severity of coronary artery disease in the Turkish population: a pilot study. Bosn J Basic Med Sci 2015; 15:9-13. [PMID: 25725138 DOI: 10.17305/bjbms.2015.157] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 11/07/2014] [Accepted: 11/07/2014] [Indexed: 11/16/2022] Open
Abstract
The cholesteryl ester transfer protein (CETP) plays a crucial role in high-density lipoprotein (HDL) metabolism. Genetic variants that alter CETP concentration may cause significant alterations in HDL-cholesterol (HDL-C) concentration. In this case-control study, we analyzed the genotype frequencies of CETP Taq1B polymorphisms in coronary artery disease patients (CAD; n=210) and controls (n=100). We analyzed the role of the CETP Taq1B variant in severity of CAD, and its association with plasma lipids and CETP concentration. DNA was extracted from 310 patients undergoing coronary angiography. The Taq1B polymorphism was genotyped using polymerase chain reaction-restriction fragment length polymorphism (RFLP) analysis. Lipid concentrations were measured by an auto analyzer and CETP level by a commercial enzyme-linked immunosorbent assay (ELISA) kit. In our study population, the B2 allele frequency was higher in control subjects than patients with single, double or triple vessel disease. B2B2 genotype carriers had a significantly higher high-density lipoprotein cholesterol (HDL-C) concentration than those with the B1B1 genotype in controls (51.93±9.47versus 45.34±9.93; p<0.05) and in CAD patients (45.52±10.81 versus 40.38±9.12; p<0.05). B2B2 genotype carriers had a significantly lower CETP concentration than those with the B1B1 genotype in controls (1.39±0.58 versus 1.88±0.83; p< 0.05) and in CAD patients (2.04±1.39versus 2.81±1.68; p< 0.05). Our data suggest that the B2 allele is associated with higher concentrations of HDL-C and lower concentrations of CETP, which confer a protective effect on coronary artery disease.
Collapse
Affiliation(s)
- Dilara Kaman
- Department of Biochemistry, Faculty of Medicine, Firat University, Elazig.
| | | | | | | |
Collapse
|
40
|
Cheng CY, Yamashiro K, Chen LJ, Ahn J, Huang L, Huang L, Cheung CMG, Miyake M, Cackett PD, Yeo IY, Laude A, Mathur R, Pang J, Sim KS, Koh AH, Chen P, Lee SY, Wong D, Chan CM, Loh BK, Sun Y, Davila S, Nakata I, Nakanishi H, Akagi-Kurashige Y, Gotoh N, Tsujikawa A, Matsuda F, Mori K, Yoneya S, Sakurada Y, Iijima H, Iida T, Honda S, Lai TYY, Tam POS, Chen H, Tang S, Ding X, Wen F, Lu F, Zhang X, Shi Y, Zhao P, Zhao B, Sang J, Gong B, Dorajoo R, Yuan JM, Koh WP, van Dam RM, Friedlander Y, Lin Y, Hibberd ML, Foo JN, Wang N, Wong CH, Tan GS, Park SJ, Bhargava M, Gopal L, Naing T, Liao J, Ong PG, Mitchell P, Zhou P, Xie X, Liang J, Mei J, Jin X, Saw SM, Ozaki M, Mizoguchi T, Kurimoto Y, Woo SJ, Chung H, Yu HG, Shin JY, Park DH, Kim IT, Chang W, Sagong M, Lee SJ, Kim HW, Lee JE, Li Y, Liu J, Teo YY, Heng CK, Lim TH, Yang SK, Song K, Vithana EN, Aung T, Bei JX, Zeng YX, Tai ES, Li XX, Yang Z, Park KH, Pang CP, Yoshimura N, Wong TY, Khor CC. New loci and coding variants confer risk for age-related macular degeneration in East Asians. Nat Commun 2015; 6:6063. [PMID: 25629512 PMCID: PMC4317498 DOI: 10.1038/ncomms7063] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 12/09/2014] [Indexed: 01/17/2023] Open
Abstract
Age-related macular degeneration (AMD) is a major cause of blindness, but presents differently in Europeans and Asians. Here, we perform a genome-wide and exome-wide association study on 2,119 patients with exudative AMD and 5,691 controls, with independent replication in 4,226 patients and 10,289 controls, all of East Asian descent, as part of The Genetics of AMD in Asians (GAMA) Consortium. We find a strong association between CETP Asp442Gly (rs2303790), an East Asian-specific mutation, and increased risk of AMD (odds ratio (OR)=1.70, P=5.60 × 10−22). The AMD risk allele (442Gly), known to protect from coronary heart disease, increases HDL cholesterol levels by 0.17 mmol l−1 (P=5.82 × 10−21) in East Asians (n=7,102). We also identify three novel AMD loci: C6orf223 Ala231Ala (OR=0.78, P=6.19 × 10−18), SLC44A4 Asp47Val (OR=1.27, P=1.08 × 10−11) and FGD6 Gln257Arg (OR=0.87, P=2.85 × 10−8). Our findings suggest that some of the genetic loci conferring AMD susceptibility in East Asians are shared with Europeans, yet AMD in East Asians may also have a distinct genetic signature. Age-related macular degeneration (AMD) is a major cause of blindness worldwide. Here, the authors carry out a two-stage genome-wide association study for AMD and identify three new AMD risk loci, highlighting the shared and distinct genetic basis of the disease in East Asians and Europeans.
Collapse
Affiliation(s)
- Ching-Yu Cheng
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Duke-NUS Graduate Medical School, National University of Singapore, Singapore 169857, Singapore [3] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore [4] Singapore National Eye Center, Singapore 168751, Singapore
| | - Kenji Yamashiro
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Li Jia Chen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jeeyun Ahn
- Department of Ophthalmology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul 156-707, Korea
| | - Lulin Huang
- 1] Sichuan Provincial Key Laboratory for Human Disease Gene Study, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China [2] School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Lvzhen Huang
- 1] Key Laboratory of Vision Loss and Restoration, Ministry of Education of China, Beijing 100044, China [2] Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing 100871, China [3] Department of Ophthalmology, People's Hospital, Peking University, Beijing 100871, China
| | - Chui Ming G Cheung
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Singapore National Eye Center, Singapore 168751, Singapore
| | - Masahiro Miyake
- 1] Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan [2] Center for Genomic Medicine/Inserm U.852, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Peter D Cackett
- 1] Singapore National Eye Center, Singapore 168751, Singapore [2] Princess Alexandra Eye Pavilion, Edinburgh EH3 9HA, UK
| | - Ian Y Yeo
- Singapore National Eye Center, Singapore 168751, Singapore
| | - Augustinus Laude
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - Ranjana Mathur
- Singapore National Eye Center, Singapore 168751, Singapore
| | - Junxiong Pang
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Kar Seng Sim
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Adrian H Koh
- 1] Singapore National Eye Center, Singapore 168751, Singapore [2] Eye and Retinal Surgeons, Camden Medical Centre, Singapore 248649, Singapore
| | - Peng Chen
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| | - Shu Yen Lee
- Singapore National Eye Center, Singapore 168751, Singapore
| | - Doric Wong
- Singapore National Eye Center, Singapore 168751, Singapore
| | - Choi Mun Chan
- Singapore National Eye Center, Singapore 168751, Singapore
| | - Boon Kwang Loh
- Singapore National Eye Center, Singapore 168751, Singapore
| | - Yaoyao Sun
- 1] Key Laboratory of Vision Loss and Restoration, Ministry of Education of China, Beijing 100044, China [2] Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing 100871, China [3] Department of Ophthalmology, People's Hospital, Peking University, Beijing 100871, China
| | - Sonia Davila
- 1] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore [2] Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Isao Nakata
- 1] Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan [2] Center for Genomic Medicine/Inserm U.852, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Hideo Nakanishi
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Yumiko Akagi-Kurashige
- 1] Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan [2] Center for Genomic Medicine/Inserm U.852, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Norimoto Gotoh
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Akitaka Tsujikawa
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine/Inserm U.852, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Keisuke Mori
- Department of Ophthalmology, Saitama Medical University, Iruma 3500495, Japan
| | - Shin Yoneya
- Department of Ophthalmology, Saitama Medical University, Iruma 3500495, Japan
| | - Yoichi Sakurada
- Department of Ophthalmology, Faculty of Medicine, University of Yamanashi, Yamanashi 4093898, Japan
| | - Hiroyuki Iijima
- Department of Ophthalmology, Faculty of Medicine, University of Yamanashi, Yamanashi 4093898, Japan
| | - Tomohiro Iida
- Department of Ophthalmology, Tokyo Women's Medical University Hospital, Tokyo 1628666, Japan
| | - Shigeru Honda
- Department of Surgery, Division of Ophthalmology, Kobe University Graduate School of Medicine, Kobe 6500017, Japan
| | - Timothy Yuk Yau Lai
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Pancy Oi Sin Tam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Haoyu Chen
- 1] Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China [2] Shantou University/Chinese University of Hong Kong Joint Shantou International Eye Center, Shantou 515041, China
| | - Shibo Tang
- 1] Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China [2] Aier School of Ophthalmology, Central South University, Changsha 410000, China
| | - Xiaoyan Ding
- Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Feng Wen
- Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Fang Lu
- 1] Sichuan Provincial Key Laboratory for Human Disease Gene Study, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China [2] School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xiongze Zhang
- Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Yi Shi
- 1] Sichuan Provincial Key Laboratory for Human Disease Gene Study, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China [2] School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Peiquan Zhao
- Department of Ophthalmology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Bowen Zhao
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Institute of Ophthalmology, Beijing 100730, China
| | - Jinghong Sang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Institute of Ophthalmology, Beijing 100730, China
| | - Bo Gong
- 1] Sichuan Provincial Key Laboratory for Human Disease Gene Study, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China [2] School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Rajkumar Dorajoo
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Jian-Min Yuan
- 1] Cancer Control and Population Sciences, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15260, USA [2] Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | - Woon-Puay Koh
- 1] Duke-NUS Graduate Medical School, National University of Singapore, Singapore 169857, Singapore [2] Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| | | | - Ying Lin
- 1] Sichuan Provincial Key Laboratory for Human Disease Gene Study, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China [2] School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Martin L Hibberd
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Jia Nee Foo
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Ningli Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Institute of Ophthalmology, Beijing 100730, China
| | - Chang Hua Wong
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Gavin S Tan
- Singapore National Eye Center, Singapore 168751, Singapore
| | - Sang Jun Park
- Department of Ophthalmology, Seoul National University Bundang Hospital, Gyeonggi 463-707, Korea
| | - Mayuri Bhargava
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore
| | - Lingam Gopal
- Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore
| | - Thet Naing
- Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore
| | - Jiemin Liao
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore
| | - Peng Guan Ong
- Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Paul Mitchell
- Department of Ophthalmology, University of Sydney and Westmead Millennium Institute, Sydney 2145, Australia
| | - Peng Zhou
- Eye and ENT Hospital of Fudan University, Shanghai 200433, China
| | | | | | - Junpu Mei
- BGI-Shenzhen, Shenzhen 518083, China
| | - Xin Jin
- BGI-Shenzhen, Shenzhen 518083, China
| | - Seang-Mei Saw
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Duke-NUS Graduate Medical School, National University of Singapore, Singapore 169857, Singapore [3] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore [4] Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| | | | | | - Yasuo Kurimoto
- Department of Ophthalmology, Kobe City General Hospital, Kobe 6500046, Japan
| | - Se Joon Woo
- 1] Department of Ophthalmology, Seoul National University Bundang Hospital, Gyeonggi 463-707, Korea [2] Department of Ophthalmology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Hum Chung
- Department of Ophthalmology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Hyeong-Gon Yu
- Department of Ophthalmology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Joo Young Shin
- Department of Ophthalmology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Dong Ho Park
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Daegu 700-721, Korea
| | - In Taek Kim
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Daegu 700-721, Korea
| | - Woohyok Chang
- Department of Ophthalmology, Yeungnam University College of Medicine, Daegu 705-802, Korea
| | - Min Sagong
- Department of Ophthalmology, Yeungnam University College of Medicine, Daegu 705-802, Korea
| | - Sang-Joon Lee
- Department of Ophthalmology, College of Medicine, Kosin University, Pusan 606-701, Korea
| | - Hyun Woong Kim
- Department of Ophthalmology, Pusan Paik Hospital, Inje University College of Medicine, Pusan 614-735, Korea
| | - Ji Eun Lee
- 1] Department of Ophthalmology, Pusan National University Hospital, Pusan 602-739, Korea [2] Medical Research Institute, Pusan National University, Pusan 602-739, Korea
| | - Yi Li
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Jianjun Liu
- 1] Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore [2] Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| | - Yik Ying Teo
- 1] Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore [2] Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| | - Chew Kiat Heng
- Department of Pediatrics, National University Health System and National University of Singapore, Singapore 119228, Singapore
| | - Tock Han Lim
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - Suk-Kyun Yang
- Department of Gastroenterology, Asan Medical Center and University of Ulsan College of Medicine, Seoul 138-736, Korea
| | - Kyuyoung Song
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 138-736, Korea
| | - Eranga N Vithana
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Duke-NUS Graduate Medical School, National University of Singapore, Singapore 169857, Singapore [3] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore
| | - Tin Aung
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore [3] Singapore National Eye Center, Singapore 168751, Singapore
| | - Jin Xin Bei
- 1] State Key Laboratory of Oncology in Southern China, Guangzhou 510060, China [2] Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou 510080, China
| | - Yi Xin Zeng
- 1] State Key Laboratory of Oncology in Southern China, Guangzhou 510060, China [2] Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou 510080, China [3] Peking Union Medical College, Chinese Academy of Medical Science, Beijing 100730, China
| | - E Shyong Tai
- 1] Duke-NUS Graduate Medical School, National University of Singapore, Singapore 169857, Singapore [2] Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore [3] Department of Medicine, National University Health System and National University of Singapore, Singapore 119228, Singapore
| | - Xiao Xin Li
- 1] Key Laboratory of Vision Loss and Restoration, Ministry of Education of China, Beijing 100044, China [2] Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing 100871, China [3] Department of Ophthalmology, People's Hospital, Peking University, Beijing 100871, China
| | - Zhenglin Yang
- 1] Sichuan Provincial Key Laboratory for Human Disease Gene Study, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China [2] School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Kyu-Hyung Park
- Department of Ophthalmology, Seoul National University Bundang Hospital, Gyeonggi 463-707, Korea
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Nagahisa Yoshimura
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Tien Yin Wong
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Duke-NUS Graduate Medical School, National University of Singapore, Singapore 169857, Singapore [3] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore [4] Singapore National Eye Center, Singapore 168751, Singapore
| | - Chiea Chuen Khor
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore [3] Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| |
Collapse
|
41
|
Mabuchi H, Nohara A, Inazu A. Cholesteryl ester transfer protein (CETP) deficiency and CETP inhibitors. Mol Cells 2014; 37:777-84. [PMID: 25410905 PMCID: PMC4255097 DOI: 10.14348/molcells.2014.0265] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 10/04/2014] [Indexed: 01/17/2023] Open
Abstract
Epidemiologic studies have shown that low-density lipoprotein cholesterol (LDL-C) is a strong risk factor, whilst high-density lipoprotein cholesterol (HDL-C) reduces the risk of coronary heart disease (CHD). Therefore, strategies to manage dyslipidemia in an effort to prevent or treat CHD have primarily attempted at decreasing LDL-C and raising HDL-C levels. Cholesteryl ester transfer protein (CETP) mediates the exchange of cholesteryl ester for triglycerides between HDL and VLDL and LDL. We have published the first report indicating that a group of Japanese patients who were lacking CETP had extremely high HDL-C levels, low LDL-C levels and a low incidence of CHD. Animal studies, as well as clinical and epidemiologic evidences, have suggested that inhibition of CETP provides an effective strategy to raise HDL-C and reduce LDL-C levels. Four CETP inhibitors have substantially increased HDL-C levels in dyslipidemic patients. This review will discuss the current status and future prospects of CETP inhibitors in the treatment of CHD. At present anacetrapib by Merck and evacetrapib by Eli Lilly are under development. By 100mg of anacetrapib HDL-C increased by 138%, and LDL-C decreased by 40%. Evacetrapib 500 mg also showed dramatic 132% increase of HDL-C, while LDL-C decreased by 40%. If larger, long-term, randomized, clinical end point trials could corroborate other findings in reducing atherosclerosis, CETP inhibitors could have a significant impact in the management of dyslipidemic CHD patients. Inhibition of CETP synthesis by antisense oligonucleotide or small molecules will produce more similar conditions to human CETP deficiency and may be effective in reducing atherosclerosis and cardiovascular events. We are expecting the final data of prospective clinical trials by CETP inhibitors in 2015.
Collapse
Affiliation(s)
- Hiroshi Mabuchi
- Department of Lipidology, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640,
Japan
| | - Atsushi Nohara
- Department of Lipidology, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640,
Japan
| | - Akihiro Inazu
- Laboratory Science, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640,
Japan
| |
Collapse
|
42
|
Corrigendum to “A potential screening factor for accumulation of cholesteyl ester transfer protein deficiency in East Asia: Schistosoma japonicum” [Biochim. Biophys. Acta 1841 (2014) 495–504]. Biochim Biophys Acta Mol Cell Biol Lipids 2014. [DOI: 10.1016/j.bbalip.2014.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
43
|
Yokoyama S. A potential screening factor for accumulation of cholesteyl ester transfer protein deficiency in East Asia: Schistosoma japonicum. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:495-504. [PMID: 24388961 DOI: 10.1016/j.bbalip.2013.12.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 12/12/2013] [Accepted: 12/24/2013] [Indexed: 11/16/2022]
Abstract
Cholesteryl ester transfer protein (CETP)-deficiency manifests a unique plasma lipoprotein profile without other apparent symptoms. It is highly common in East Asia while rather rare anywhere else. A potential environmental screening factor(s) may therefore contribute to this eccentric distribution, such as its selective advantage against a regional illness, most likely an infectious disease, in relation to plasma lipoproteins. Blood flukes use the host plasma lipoproteins as nutrient sources through the lipoprotein receptor-like systems. Its Asian-specific species, Schistosoma (S) japonicum, which has been endemic in East Asia, takes up cholesteryl ester (CE) from high-density lipoprotein (HDL) for the embryonation of their eggs to miracidia, a critical step of the hepatic pathogenesis of this parasite, but poorly from HDL of CETP-deficiency. CD36-related protein (CD36RP) was cloned from the adults and the eggs of S. japonicum, with 1880-bp encoding 506 amino-acid residues exhibiting the CD36 domains and two transmembrane regions. Its extracellular domain selectively bound human HDL but neither LDL nor CETP-deficiency HDL, and the antibody against the extracellular domain suppressed the selective HDL-CE uptake and embryonation of the eggs. When infected with S. japonicum, wild-type mice developed less hepatic granulomatosis than CETP-transgenic mice by the ectopic egg embryonation. CD36RP is thus a candidate receptor of S. japonicum to facilitate uptake of HDL-CE necessary for egg embryonation. Abnormal HDL caused by CETP-deficiency retards this process and thereby protects the patients from development of hepatic lesions. S. japonicum infection is a potential screening factor for high prevalence of CETP deficiency in East Asia.
Collapse
Affiliation(s)
- Shinji Yokoyama
- Nutritional Health Science Research Center, Chubu University, Matsumoto-cho 1200, Kasugai 487-8501, Japan.
| |
Collapse
|
44
|
de Almeida ERD, Reiche EMV, Kallaur AP, Flauzino T, Watanabe MAE. The roles of genetic polymorphisms and human immunodeficiency virus infection in lipid metabolism. BIOMED RESEARCH INTERNATIONAL 2013; 2013:836790. [PMID: 24319689 PMCID: PMC3844249 DOI: 10.1155/2013/836790] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 09/02/2013] [Accepted: 09/11/2013] [Indexed: 12/31/2022]
Abstract
Dyslipidemia has been frequently observed among individuals infected with human immunodeficiency virus type 1 (HIV-1), and factors related to HIV-1, the host, and antiretroviral therapy (ART) are involved in this phenomenon. This study reviews the roles of genetic polymorphisms, HIV-1 infection, and highly active antiretroviral therapy (HAART) in lipid metabolism. Lipid abnormalities can vary according to the HAART regimen, such as those with protease inhibitors (PIs). However, genetic factors may also be involved in dyslipidemia because not all patients receiving the same HAART regimen and with comparable demographic, virological, and immunological characteristics develop variations in the lipid profile. Polymorphisms in a large number of genes are involved in the synthesis of structural proteins, and enzymes related to lipid metabolism account for variations in the lipid profile of each individual. As some genetic polymorphisms may cause dyslipidemia, these allele variants should be investigated in HIV-1-infected patients to identify individuals with an increased risk of developing dyslipidemia during treatment with HAART, particularly during therapy with PIs. This knowledge may guide individualized treatment decisions and lead to the development of new therapeutic targets for the treatment of dyslipidemia in these patients.
Collapse
Affiliation(s)
- Elaine Regina Delicato de Almeida
- Department of Pathology, Clinical Analysis and Toxicology, Health Sciences Center, State University of Londrina, Avenida Robert Koch, 60, CEP 86038-440 Londrina, PR, Brazil
- Pathological Sciences Postgraduate Program, Biological Sciences Center, State University of Londrina, Campus Universitário, CEP 86051-970 Londrina, PR, Brazil
| | - Edna Maria Vissoci Reiche
- Department of Pathology, Clinical Analysis and Toxicology, Health Sciences Center, State University of Londrina, Avenida Robert Koch, 60, CEP 86038-440 Londrina, PR, Brazil
| | - Ana Paula Kallaur
- Postgraduate Program of Health Sciences Center, State University of Londrina, Avenida Robert Koch, 60, CEP 86038-440 Londrina, PR, Brazil
| | - Tamires Flauzino
- Clinical Immunology, Clinical Analysis Laboratory, Health Sciences Center, State University of Londrina, Avenida Robert Koch, 60, CEP 86038-440 Londrina, PR, Brazil
| | - Maria Angelica Ehara Watanabe
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Campus Universitário, CEP 86051-970 Londrina, PR, Brazil
| |
Collapse
|
45
|
|
46
|
Teramoto T, Sasaki J, Ishibashi S, Birou S, Daida H, Dohi S, Egusa G, Hiro T, Hirobe K, Iida M, Kihara S, Kinoshita M, Maruyama C, Ohta T, Okamura T, Yamashita S, Yokode M, Yokote K. Cardiovascular disease risk factors other than dyslipidemia. Executive summary of the Japan Atherosclerosis Society (JAS) guidelines for the diagnosis and prevention of atherosclerotic cardiovascular diseases in Japan - 2012 version. J Atheroscler Thromb 2013; 20:733-42. [PMID: 23892529 DOI: 10.5551/jat.17368] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Tamio Teramoto
- Committee for Epidemiology and Clinical Management of Atherosclerosis
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Guay SP, Brisson D, Lamarche B, Marceau P, Vohl MC, Gaudet D, Bouchard L. DNA methylation variations at CETP and LPL gene promoter loci: new molecular biomarkers associated with blood lipid profile variability. Atherosclerosis 2013; 228:413-20. [PMID: 23623643 DOI: 10.1016/j.atherosclerosis.2013.03.033] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 02/16/2013] [Accepted: 03/09/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND Recent findings suggest that DNA methylation, a well-known epigenetic mechanism, is involved in high-density lipoprotein cholesterol (HDL-C) metabolism and increased cardiovascular disease risk. The aim of this study was thus to assess whether DNA methylation within key genes of lipoprotein metabolism is associated with blood lipid profile variability. METHODS AND RESULTS Ninety-eight untreated familial hypercholesterolaemia patients (61 men and 37 women) were recruited for leucocyte DNA methylation analyses at the LDLR, CETP, LCAT and LPL gene promoter loci using bisulfite pyrosequencing. LPL DNA methylation was correlated with HDL-C (r = 0.22; p = 0.031) and HDL particle size (r = 0.47, p = 0.013). In both sex, CETP DNA methylation was negatively associated with low-density lipoprotein cholesterol levels (r < -0.32; p < 0.05). In men, CETP DNA methylation was associated with HDL-C (r = -0.36; p = 0.006), HDL-triglyceride levels (r = 0.59; p < 0.001) and HDL particle size (r = -0.44, p = 0.019). In visceral adipose tissue from 30 men with severe obesity, the associations between LPL DNA methylation, HDL-C (r = -0.40; p = 0.03) and LPL mRNA levels (r = -0.61, p < 0.001) were confirmed. CONCLUSION CETP and LPL DNA methylation levels are associated with blood lipid profile, suggesting that further studies of epipolymorphisms should most certainly contribute to a better understanding of the molecular bases of dyslipidemia.
Collapse
Affiliation(s)
- S P Guay
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | | | | | | | | | | | | |
Collapse
|
48
|
Rosenson RS, Brewer HB, Davidson WS, Fayad ZA, Fuster V, Goldstein J, Hellerstein M, Jiang XC, Phillips MC, Rader DJ, Remaley AT, Rothblat GH, Tall AR, Yvan-Charvet L. Cholesterol efflux and atheroprotection: advancing the concept of reverse cholesterol transport. Circulation 2012; 125:1905-19. [PMID: 22508840 PMCID: PMC4159082 DOI: 10.1161/circulationaha.111.066589] [Citation(s) in RCA: 716] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Robert S Rosenson
- Mount Sinai Heart, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ohtani R, Inazu A, Noji Y, Wakasugi T, Miwa K, Tada H, Kawashiri MA, Noguchi T, Nohara A, Kobayashi J, Koizumi J, Yamagishi M, Mabuchi H. Novel mutations of cholesteryl ester transfer protein (CETP) gene in Japanese hyperalphalipoproteinemic subjects. Clin Chim Acta 2012; 413:537-43. [DOI: 10.1016/j.cca.2011.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 11/08/2011] [Indexed: 10/15/2022]
|
50
|
Hossain MA, Ngeth S, Chan T, Oda MN, Francis GA. Lipid-bound apolipoproteins in tyrosyl radical-oxidized HDL stabilize ABCA1 like lipid-free apolipoprotein A-I. BMC BIOCHEMISTRY 2012; 13:1. [PMID: 22248050 PMCID: PMC3292454 DOI: 10.1186/1471-2091-13-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 01/16/2012] [Indexed: 11/20/2022]
Abstract
Background ATP-binding cassette transporter A1 (ABCA1) mediates the lipidation of exchangeable apolipoproteins, the rate-limiting step in the formation of high density lipoproteins (HDL). We previously demonstrated that HDL oxidized ex vivo by peroxidase-generated tyrosyl radical (tyrosylated HDL, tyrHDL) increases the availability of cellular cholesterol for efflux and reduces the development of atherosclerosis when administered to apolipoprotein E-deficient mice as compared to treatment with control HDL. Results In the current study we determined that tyrHDL requires functional ABCA1 for this enhanced activity. Like lipid-free apolipoprotein A-I (apoA-I), tyrHDL increases total and cell surface ABCA1, inhibits calpain-dependent and -independent proteolysis of ABCA1, and can be bound by cell surface ABCA1 in human skin fibroblasts. Additionally, tyrHDL apoproteins are susceptible to digestion by enteropeptidase like lipid-free apoA-I, but unlike lipid-bound apoA-I on HDL, which is resistant to proteolysis. Conclusions These results provide the first evidence that lipid-bound apolipoproteins on the surface of spherical HDL particles can behave like lipid-free apoA-I to increase ABCA1 protein levels and activity.
Collapse
Affiliation(s)
- Mohammad A Hossain
- Department of Medicine, UBC James Hogg Research Centre, Heart and Lung Institute, St, Paul's Hospital, Vancouver, British Columbia, Canada
| | | | | | | | | |
Collapse
|