Skip to main content
Drugs in Context logoLink to Drugs in Context
. 2018 Mar 28;7:212525. doi: 10.7573/dic.212525

High-density lipoprotein (HDL) functionality and its relevance to atherosclerotic cardiovascular disease

Constantine E Kosmas 1,, Ian Martinez 2, Andreas Sourlas 3, Kyriaki V Bouza 4, Frederick N Campos 2, Verenisse Torres 2, Peter D Montan 2, Eliscer Guzman 5
PMCID: PMC5877920  PMID: 29623098

Abstract

Several prospective epidemiological studies have shown that there is a clear inverse relationship between serum high-density lipoprotein-cholesterol (HDL-C) concentrations and risk for coronary heart disease (CHD), even at low-density lipoprotein-cholesterol (LDL-C) levels below 70 mg/dL. However, more recent evidence from genetic studies and clinical research has come to challenge the long-standing notion that higher HDL-C levels are always beneficial, while lower HDL-C levels are always detrimental. Thus, it becomes apparent that HDL functionality plays a much more important role in atheroprotection than circulating HDL-C levels. HDL cholesterol efflux capacity (CEC) from macrophages is a key metric of HDL functionality and exhibits a strong inverse association with both carotid intima-media thickness and the likelihood of angiographic coronary artery disease (CAD), independent of the HDL-C level. Thus, extensive research is being conducted to identify new agents with a favorable side effect profile, which would be able to enhance CEC, improve HDL functionality and potentially decrease cardiovascular risk. This review aims to present and discuss the current clinical and scientific evidence pertaining to the significance of HDL functionality over the actual HDL-C concentration in mediating the favorable effects on the cardiovascular system. Thus, we conducted a PubMed search until December 2017 through the English literature using the search terms ‘HDL function/functionality’, ‘HDL properties’, ‘cardiovascular risk’ and ‘cholesterol efflux capacity’. We also included references from the articles identified and publications available in the authors’ libraries.

Keywords: ApoA-I Milano, cardiovascular disease (CVD), cholesterol efflux capacity (CEC), HDL-cholesterol (HDL-C), HDL functionality

Introduction

Cardiovascular disease (CVD) is the leading cause of death worldwide, being responsible for approximately 30% of the annual global mortality [1]. In the United States the disease is highly prevalent and over one-third of the population has CVD [2]. Several prospective epidemiological studies have shown that there is a clear inverse relationship between serum high-density lipoprotein-cholesterol (HDL-C) concentrations and risk for coronary heart disease (CHD), even at low-density lipoprotein-cholesterol (LDL-C) levels below 70 mg/dL [3,4]. Furthermore, it has been estimated that for each increment of 1 mg/dL in HDL-C, the CHD risk is reduced by 3% in women and by 2% in men [5].

However, more recent evidence from genetic studies and clinical research has come to challenge the long-standing notion that higher HDL-C levels are invariably beneficial, while lower HDL-C levels are always detrimental [6].

In a genetic study, three functional variants of hepatic lipase associated with a modest rise in levels of HDL-C did not improve cardiovascular risk [7]. On the other hand, functional mutations in ATP-binding cassette transporter A1 (ABCA1) leading to a 29.3% reduction in HDL-C levels, did not adversely affect cardiovascular risk [8]. Furthermore, it was shown that carriers of a single nucleotide polymorphism in the endothelial lipase gene leading to an increase of HDL-C levels by 5.4 mg/dL, but with similar levels of other lipid and nonlipid cardiovascular risk factors, did not enjoy a reduced risk of myocardial infarction, as compared with noncarriers [9]. In a review, which discussed the latest insights for HDL-C-associated genes, as well as evidence from large-scale genome-wide association studies, it was concluded that the current data do not support a direct causal relation between molecularly defined HDL disorders and atherosclerosis per se [6,10]. In a large clinical study, higher preoperative HDL-C levels were not associated with reduced risk of vascular events in patients with coronary artery disease (CAD) undergoing coronary artery bypass grafting (CABG) [11]. More importantly, it has been shown that very high LDL-C levels and/or very large HDL particle size are associated with increased risk for CAD [12]. This observation, along with the detrimental, neutral or at most slightly positive effects on CVD risk conferred by the cholesteryl ester transfer protein (CETP) inhibitors, despite a large increase in HDL-C levels (from 31% and up to 133.2%) [1316], significantly weaken the notion that high HDL-C levels are always protective [6].

This review aims to present and discuss the current clinical and scientific evidence pertaining to the significance of HDL functionality over the actual HDL-C concentration for determining cardiovascular risk. Thus, we conducted a PubMed search until December 2017 through the English literature using the search terms ‘HDL function/functionality’, ‘HDL properties’, ‘cardiovascular risk’ and ‘cholesterol efflux capacity’. We also included references from the articles identified and publications available in the authors’ libraries.

Composition and biological functionality of HDL

HDL constitutes a heterogeneous group of particles differing in density, size, electrophoretic mobility, and apolipoprotein content [17]. The major HDL apolipoproteins are ApoA-I and ApoA-II, and both are required for normal HDL biosynthesis. ApoA-I is synthesized in both the intestine and the liver, constitutes approximately 70% of HDL protein, and is present on virtually all HDL particles. ApoA-II is synthesized only in the liver, constitutes approximately 20% of HDL protein, and is present on about two-thirds of HDL particles in humans [18].

Mass spectrometry studies have revealed that the HDL particles carry a multiplicity of proteins, which not only affect lipid metabolism but are also involved in complement regulation, acute-phase response and proteinase inhibition [19]. Lipidomic approaches have identified more than 200 molecular lipid species in normolipidemic HDL, including phospholipids, sphingolipids, steroids, cholesteryl esters, triglycerides, diacylglycerides, monoacylglycerides and free fatty acids [20]. Furthermore, it has to be emphasized that HDL consists of a group of particles with marked structural, physiochemical, compositional and functional heterogeneity and with significant differences in their biological activities [6,20,21]. Thus, it becomes evident that, given this vast heterogeneity of biological functions, HDL functionality cannot be inferred from the plain measurement of plasma HDL-C levels [22].

HDL plays a major role in reverse cholesterol transport (RCT), by which excess cholesterol is removed from the peripheral vessels and is transported back to the liver for disposal [23]. However, HDL has several other beneficial biological properties, which enhance its protective effect against CVD. These include antioxidative, anti-inflammatory endothelial/vasodilatory, antithrombotic and cytoprotective functions [2427]. More specifically, HDL may provide potent protection of LDL in vivo from oxidative damage, induced by free radicals in the arterial intima, with consequent inhibition of the generation of proinflammatory oxidized lipids, mainly lipid hydroperoxides but also short-chain oxidized phospholipids [24]. HDL also inhibits the expression of adhesion molecules in endothelial cells and thus it decreases the recruitment of blood monocytes into the arterial wall [25]. HDL also increases the production of the atheroprotective signaling molecule nitric oxide (NO) via upregulation of the expression of endothelial NO synthase (eNOS), as well as by maintaining the lipid environment in caveolae, where eNOS is co-localized with partner signaling molecules. In addition, HDL stimulates eNOS as a result of kinase cascade activation by the high-affinity HDL receptor, scavenger receptor class B type I (SR-BI) [26]. The antithrombotic function of HDL may be exerted through the activation of prostacyclin synthesis, as well as through the attenuation of the expression of tissue factor and selectins, with consequent downregulation of thrombin generation via the protein C pathway and direct and indirect blunting of platelet activation [26]. The direct cytoprotective effect of HDL on endothelial cells may be exerted through prevention of the suicide pathway leading to apoptosis of endothelial cells by decreasing the cysteine protease P32 (CPP32)-like protease activity. Thus, HDL plays a protective role against ‘injury’, as this is described in the ‘response-to-injury’ hypothesis of atherogenesis [27].

The ApoA-I Milano mutation: low levels of a highly functional HDL offering protection from CVD

The ApoA-I Milano (AI-M) mutation was first described in 1980 in a family originating from Limone sul Garda, a small town outside Milan in northern Italy. In this genetic mutation, the ApoA-I variant shows a single amino acid substitution of arginine to cysteine at the position 173 in the primary sequence of ApoA-I. This substitution leads to the formation of homodimers (AI-M/AI-M) and heterodimers with apolipoprotein AII (AI-M/AII). The carriers of the ApoA-I Milano mutation share a lipid profile characterized by very low HDL-C levels and moderate hypertriglyceridemia but without evidence of premature CAD or preclinical coronary or carotid atherosclerosis [28,29].

Weekly infusions of recombinant ApoA-I Milano, as compared with placebo, caused a significant regression of coronary atherosclerosis in patients with acute coronary syndrome (ACS) after 5 only treatments [30]. Furthermore, in an animal study, recombinant ApoA-1 Milano was shown to exert greater anti-inflammatory, antioxidant and plaque-stabilizing effects, as compared with wild-type HDL [31].

These cardiovascular protective effects of ApoA-I Milano provided the initial clinical support for the concept that the functionality of HDL plays a more important role to reduce atherosclerosis than the circulating level of HDL-C [32].

HDL particle subpopulations and HDL functionality

As it was alluded before, plasma HDL constitutes a heterogeneous group of particles with diverse structure and biological activity, mainly due to differences in their lipid and apolipoprotein content [6]. Earlier studies have indicated that the larger HDL particles are more protective [3336]. However, more recent evidence has come to challenge this concept.

CETP inhibitors failed to provide any meaningful reduction in cardiovascular risk despite a substantial increase in circulating HDL-C levels [1316]. Similarly, niacin (added to statin therapy) also failed to decrease cardiovascular risk despite a significant increase in HDL-C levels [37,38]. Both CETP inhibitors and niacin preferentially increase the levels of the large HDL particles, whereas their effects on the total number of HDL particles are weaker [39]. There is evidence from experimental studies that cholesterol-overloaded HDL particles may be functionally abnormal with impaired anti-atherogenic potential; they may have a negative impact on the efflux potential of cholesterol from extrahepatic cells and may reduce hepatic selective uptake of cholesterol mediated by scavenger receptor SR-BI [40,41]. This evidence is also supported by a post hoc analysis of two large prospective studies, the IDEAL (Incremental Decrease in End Points through Aggressive Lipid Lowering) trial and the EPIC (European Prospective Investigation into Cancer and Nutrition)-Norfolk case-control study, which showed that very high plasma HDL-C levels (≥70 mg/dL) and very large HDL particles (>9.53 nm) were associated with higher risk for CVD. In contrast, ApoA-I remained protective across the major part of its distribution in both studies, thus proving that high plasma ApoA-I more uniformly represents lower risk [12]. Furthermore, in another community-based cohort study, it was clearly shown that cholesterol-overloaded HDL particles were independently associated with progression of carotid atherosclerosis in a CVD-free population. More specifically, participants with the highest estimated number of cholesterol molecules per HDL particle (≥53.0) had 1.56-fold (95% confidence interval: 1.14 to 2.13; p=0.006) increased progression, as compared with those with the lowest estimated number of cholesterol molecules per HDL particle (<41.0) [41].

On the other hand, there is evidence showing that small-dense HDL particles may be more ‘functional’ in many protective mechanisms. More specifically, small-dense HDL particles promote more effectively cholesterol efflux from lipid-loaded macrophages and exhibit more potent antioxidative, anti-inflammatory, cytoprotective, antithrombotic and anti-infectious activity, as compared with larger HDL particles [6,22,42].

The apparent discordancy concerning the atheroprotective potential of the different subfractions of the HDL particles may well be elucidated by more recent data showing that the presence of large HDL particles is linked to a lower number of circulating LDL particles, and primarily of the highly atherogenic small-dense LDL particles [43]. Therefore, it becomes apparent that the cardiovascular protection may not be related to the large size of HDL particles but may be actually due to the associated reduced number of LDL particles [6].

In a large multiethnic study of patients without baseline CHD, which was designed to evaluate the independent associations of HDL-C and HDL particle (HDL-P) concentrations with carotid intima-media thickness (CIMT) and incident CHD, it was clearly shown that, after adjusting for each other and LDL particle (LDL-P) concentration, the concentration of HDL-C was no longer associated with CIMT or CHD, whereas HDL-P remained independently associated with both CIMT and CHD [44]. These data again clearly support the concept that the anti-atherogenic potential of HDL may be related to the total HDL-P concentration but cannot be inferred from the plain measurement of plasma HDL-C.

Factors altering HDL functionality

As mentioned above, HDL plays a major role in RCT, but also exhibits antioxidative, anti-inflammatory endothelial/vasodilatory, antithrombotic and cytoprotective functions. On the other hand, the major proteins of HDL are ApoA-I and ApoA-II but HDL particles also carry a multiplicity of less abundant proteins, which not only affect lipid metabolism but are also involved in complement regulation, acute-phase response and proteinase inhibition. These include ApoC-I, ApoC-II, apoC-III, apoE, apoJ, apoL, lecithin:cholesterol acyl-transferase (LCAT), serum paraoxonase-1 (PON1), and platelet-activating factor acetylhydrolase (PAF-AH) [45]. Modification of the protein components of HDL, brought about by the oxidative environment of the acute-phase response (a systemic response to infection, surgery, myocardial infarction, and chronic inflammation), can convert HDL from an anti-inflammatory to a proinflammatory particle [45,46]. Indeed, during acute-phase response, a new set of proteins, including serum amyloid A (SAA) and ceruloplasmin, bind to HDL and may render HDL pro-inflammatory and pro-atherogenic by limiting its ability to promote RCT and to prevent LDL modification [45]. In a study using human aortic endothelial and smooth muscle cells, it was shown that while HDL obtained before cardiac surgery completely inhibited the LDL-induced increase in monocyte transmigration, in contrast, acute-phase HDL, obtained from the same patients 2–3 days after surgery, not only did not inhibit the LDL-induced increase in monocyte transmigration but also amplified it by up to 1.8-fold (p<0.01). Thus, anti-inflammatory HDL actually became pro-inflammatory during the acute-phase response [47].

Furthermore, recent studies have shown that HDL and ApoA-I recovered from human atheroma are dysfunctional and are extensively oxidized by myeloperoxidase (MPO). More specifically, ApoA-I containing a 2-OH-Trp72 group (oxTrp72-apoA1) is in low abundance within the circulation but accounts for 20% of the apoA1 in atherosclerosis-laden arteries. Moreover, increased levels of oxTrp72-apoA1 in subjects presenting to a cardiology clinic were associated with increased risk for CVD. Thus, it was suggested that circulating levels of oxTrp72-apoA1 may serve as a marker of the pro-atherogenic process in the arterial wall [48]. In accordance with this study, there is evidence indicating that dysfunctional HDLs have impaired anti-inflammatory potential and their presence in patients with CAD may actually help discriminate between patients with ACS and patients with stable CAD [49].

In addition, it has been shown that glycation may also impair HDL function and this could be a contributing factor to the accelerated atherosclerosis observed in Type II diabetes mellitus [50]. Furthermore, there is evidence that in metabolic syndrome the small-dense HDL particles become dysfunctional displaying impaired antioxidative activity [51]. Moreover, protein carbamylation may also render HDL dysfunctional, which raises the possibility that HDL carbamylation contributes to foam cell formation in atherosclerotic lesions [52]. Additionally, MPO-mediated oxidation of ApoA-I has also been found to impair HDL function in regard to its RCT, antioxidant and anti-inflammatory activities [53].

Ethnicity has also been shown to be associated with HDL functionality. In this regard, there is evidence that black South African women, in comparison to white women, display improved HDL antioxidant functionality and are relatively protected against CHD despite greater prevalence of obesity and lower circulating HDL-C levels than white women [54]. There is also evidence that obesity may impair HDL functionality [54,55], whereas bariatric surgery may actually exert a significant improvement in HDL structure and functionality [56].

Dietary habits may also play a significant role in the functionality of HDL. Consumption of saturated fat has been shown to reduce the anti-inflammatory potential of HDL and impair arterial endothelial function. In contrast, consumption of polyunsaturated fat is associated with an improvement of the anti-inflammatory activity of HDL. These findings highlight novel mechanisms by which different dietary fatty acids may affect atherogenicity [57].

HDL cholesterol efflux capacity: a key metric of HDL functionality

Cholesterol efflux from macrophages to HDL occurs via several mechanisms involving the ABCA1 and ABCG1 transporters, as well as the scavenger receptor SR-BI [58]. Although cholesterol efflux from macrophages accounts only for a very small fraction of the total efflux of cholesterol from peripheral tissues via the RCT pathway, it appears to be the most relevant component in regard to atheroprotection and therefore it may provide an excellent surrogate for HDL functionality [6,59]. Actually, in a cross-sectional study, it was demonstrated that the cholesterol efflux capacity (CEC) from macrophages has a potent inverse association with both CIMT and the likelihood of angiographic CAD, independent of the HDL-C level [60]. The results of this study were confirmed in another recent study, which followed 2,924 adults free from cardiovascular disease who were participants in the Dallas Heart Study over a median follow-up period of 9.4 years. HDL-C level, HDL-P concentration and CEC were measured at baseline. There was a statistically significant 67% reduction in cardiovascular risk in the highest quartile of CEC compared with the lowest quartile. Adding CEC to traditional risk factors was associated with improvement in discrimination and reclassification indexes [61].

In addition, there is evidence that CEC is impaired in patients with chronic kidney disease, metabolic syndrome, diabetes mellitus and autoimmune disorders [62]. On the other hand, CEC is actually enhanced in patients with the metabolic syndrome and low HDL-C levels who were treated with pioglitazone, but not in patients with hypercholesterolemia who were treated with statins [60]. It has also been shown that male sex and current smoking are associated with decreased CEC [60].

Thus, quantification of CEC may be instrumental in the assessment of new therapies targeting HDL metabolism and RCT, as agents that enhance CEC may lead to improvement of HDL functionality and potentially reduction of cardiovascular risk.

Future therapeutic directions

As it was mentioned earlier in this review, infusions of recombinant ApoA-I Milano caused a significant regression of coronary atherosclerosis in patients with ACS [30]. However, subsequent clinical development was delayed by several years due to manufacturing difficulties and contamination from host-derived proteins [32]. More recently, a clean manufacturing process was developed to produce the recombinant ApoA-I Milano without contamination by host-derived proteins and this new product was called MDCO-216 [32]. However, in a pilot trial, MDCO-216 did not produce a significant beneficial effect on CAD progression measured by Intravascular Ultrasound (IVUS) [63] and the sponsor company abandoned its further development.

It is known that plasma-selective delipidation converts αHDL to preβ-like HDL, the most effective form of HDL for lipid removal from arterial plaques [64]. Autologous delipidated HDL plasma infusions in patients with ACS were proven to be clinically feasible and well tolerated. Furthermore, the IVUS data demonstrated a numeric trend toward regression in the total atheroma volume in the delipidated group compared with an increase of total atheroma volume in the control group, although the results did not reach statistical significance [64]. Further studies will be needed to determine the ability of this therapy to reduce clinical cardiovascular events.

In a randomized, double-blind, placebo-controlled, dose-ranging phase 2b trial, which was designed to assess the safety and tolerability of CSL112 (a reconstituted, infusible, plasma-derived ApoA-I) after acute myocardial infarction (AEGIS-I trial), it was shown that 4 weekly infusions of CSL112 were feasible, well tolerated, and not associated with any significant impairment in liver or kidney function or other safety concerns. The ability of CSL112 to acutely enhance CEC was also confirmed [65]. A phase 3 trial to assess the potential benefit of CSL112 to reduce major adverse cardiovascular events has already been planned and will begin recruiting patients in early 2018.

Although infusions of recombinant ApoA-I Milano or ApoA-I wild type may be potentially safe and effective for clinical application, this therapy is limited for clinical use due to the high cost of large-scale production of ApoA-I and need for repeated intravenous administration. Thus, gene therapy could represent an alternative approach for its possible long-term effect. Although preclinical studies have provided some evidence of benefit using this approach, further studies are needed for its definitive clinical validation [32]. Recent progress in recombinant adeno-associated virus (AAV) technology appears promising in this regard [32,66].

Conclusions

From the above review of the scientific, epidemiological and clinical data, it becomes apparent that HDL functionality plays a much more important role in atheroprotection than circulating HDL-C levels. Plasma HDL constitutes a heterogeneous group of particles with diverse structure and biological activity, and very high HDL-C levels are not invariably protective but rather, under certain conditions, may actually become pro-inflammatory. HDL functionality is dependent upon genetic, environmental, and lifestyle factors and may be modified in several disease states. HDL CEC from macrophages is a key metric of HDL functionality and exhibits a strong inverse association with both CIMT and the likelihood of angiographic CAD, independent of the HDL-C level. Thus, extensive research is being conducted to identify new agents with a favorable side-effect profile, which would be able to enhance CEC, improve HDL functionality and potentially decrease cardiovascular risk.

Footnotes

Disclosure and potential conflicts of interest: Constantine E Kosmas is a member of the Dyslipidemia Speaker Bureau of Amgen, Inc. The International Committee of Medical Journal Editors (ICMJE) Potential Conflicts of Interests form for the authors are available for download at: http://www.drugsincontext.com/wp-content/uploads/2018/03/dic.212525-COI.pdf

Correct attribution: Copyright © 2018 Kosmas CE, Martinez I, Sourlas A, Bouza KV, Campos FN, Torres V, Montan PD, Guzman E. https://doi.org/10.7573/dic.212525. Published by Drugs in Context under Creative Commons License Deed CC BY NC ND 4.0.

Provenance: invited; externally peer reviewed.

Peer review comments to author: 01 March 2018

Drugs in Context is published by BioExcel Publishing Ltd. Registered office: Plaza Building, Lee High Road, London, England, SE13 5PT.

BioExcel Publishing Limited is registered in England Number 10038393. VAT GB 252772009.

For all manuscript and submissions enquiries, contact the Editorial office dic.editorial@bioexcelpublishing.com

For all permissions, rights and reprints, contact David Hughes david.hughes@bioexcelpublishing.com

References

  • 1.Roth GA, Johnson C, Abajobir A, Abd-Allah F, Abera SF, Abyu G, Ahmed M, Aksut B, Alam T, Alam K, Alla F, Alvis-Guzman N, Amrock S, Ansari H, Ärnlöv J, Asayesh H, Atey TM, Avila-Burgos L, Awasthi A, Banerjee A, Barac A, Bärnighausen T, Barregard L, Bedi N, Belay Ketema E, Bennett D, Berhe G, Bhutta Z, Bitew S, Carapetis J, Carrero JJ, Malta DC, Castañeda-Orjuela CA, Castillo-Rivas J, Catalá-López F, Choi JY, Christensen H, Cirillo M, Cooper L, Jr, Criqui M, Cundiff D, Damasceno A, Dandona L, Dandona R, Davletov K, Dharmaratne S, Dorairaj P, Dubey M, Ehrenkranz R, El Sayed Zaki M, Faraon EJA, Esteghamati A, Farid T, Farvid M, Feigin V, Ding EL, Fowkes G, Gebrehiwot T, Gillum R, Gold A, Gona P, Gupta R, Habtewold TD, Hafezi-Nejad N, Hailu T, Hailu GB, Hankey G, Hassen HY, Abate KH, Havmoeller R, Hay SI, Horino M, Hotez PJ, Jacobsen K, James S, Javanbakht M, Jeemon P, John D, Jonas J, Kalkonde Y, Karimkhani C, Kasaeian A, Khader Y, Khan A, Khang YH, Khera S, Khoja AT, Khubchandani J, Kim D, Kolte D, Kosen S, Krohn KJ, Kumar GA, Kwan GF, Lal DK, Larsson A, Linn S, Lopez A, Lotufo PA, El Razek HMA, Malekzadeh R, Mazidi M, Meier T, Meles KG, Mensah G, Meretoja A, Mezgebe H, Miller T, Mirrakhimov E, Mohammed S, Moran AE, Musa KI, Narula J, Neal B, Ngalesoni F, Nguyen G, Obermeyer CM, Owolabi M, Patton G, Pedro J, Qato D, Qorbani M, Rahimi K, Rai RK, Rawaf S, Ribeiro A, Safiri S, Salomon JA, Santos I, Santric Milicevic M, Sartorius B, Schutte A, Sepanlou S, Shaikh MA, Shin MJ, Shishehbor M, Shore H, Silva DAS, Sobngwi E, Stranges S, Swaminathan S, Tabarés-Seisdedos R, Tadele Atnafu N, Tesfay F, Thakur JS, Thrift A, Topor-Madry R, Truelsen T, Tyrovolas S, Ukwaja KN, Uthman O, Vasankari T, Vlassov V, Vollset SE, Wakayo T, Watkins D, Weintraub R, Werdecker A, Westerman R, Wiysonge CS, Wolfe C, Workicho A, Xu G, Yano Y, Yip P, Yonemoto N, Younis M, Yu C, Vos T, Naghavi M, Murray C. Global, Regional, and National Burden of Cardiovascular Diseases for 10 Causes, 1990 to 2015. J Am Coll Cardiol. 2017;70(1):1–25. doi: 10.1016/j.jacc.2017.04.052. http://dx.doi.org/10.1016/j.jacc.2017.04.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Writing Group Members; Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, Das SR, de Ferranti S, Després JP, Fullerton HJ, Howard VJ, Huffman MD, Isasi CR, Jiménez MC, Judd SE, Kissela BM, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Magid DJ, McGuire DK, Mohler ER, 3rd, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Rosamond W, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Woo D, Yeh RW, Turner MB American Heart Association Statistics Committee; Stroke Statistics Subcommittee. Heart disease and stroke statistics-2016 update: a report from the American Heart Association. Circulation. 2016;133(4):e38–360. doi: 10.1161/CIR.0000000000000350. http://dx.doi.org/10.1161/CIR.0000000000000350. [DOI] [PubMed] [Google Scholar]
  • 3.Castelli WP. Cholesterol and lipids in the risk of coronary artery disease--the Framingham Heart Study. Can J Cardiol. 1988;4(Suppl A):5A–10A. [PubMed] [Google Scholar]
  • 4.Barter P, Gotto AM, LaRosa JC, Maroni J, Szarek M, Grundy SM, Kastelein JJ, Bittner V, Fruchart JC Treating to New Targets Investigators. HDL cholesterol, very low levels of LDL cholesterol, and cardiovascular events. N Engl J Med. 2007;357(13):1301–10. doi: 10.1056/NEJMoa064278. http://dx.doi.org/10.1056/NEJMoa064278. [DOI] [PubMed] [Google Scholar]
  • 5.Gordon DJ, Probstfield JL, Garrison RJ, Neaton JD, Castelli WP, Knoke JD, Jacobs DR, Jr, Bangdiwala S, Tyroler HA. High-density lipoprotein cholesterol and cardiovascular disease. Four prospective American studies. Circulation. 1989;79(1):8–15. doi: 10.1161/01.cir.79.1.8. http://dx.doi.org/10.1161/01.CIR.79.1.8. [DOI] [PubMed] [Google Scholar]
  • 6.Kosmas CE, Christodoulidis G, Cheng JW, Vittorio TJ, Lerakis S. High-density lipoprotein functionality in coronary artery disease. Am J Med Sci. 2014;347(6):504–8. doi: 10.1097/MAJ.0000000000000231. http://dx.doi.org/10.1097/MAJ.0000000000000231. [DOI] [PubMed] [Google Scholar]
  • 7.Vergeer M, Holleboom AG, Kastelein JJ, Kuivenhoven JA. The HDL hypothesis: does high-density lipoprotein protect from atherosclerosis? J Lipid Res. 2010;51(8):2058–73. doi: 10.1194/jlr.R001610. http://dx.doi.org/10.1194/jlr.R001610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Frikke-Schmidt R, Nordestgaard BG, Stene MC, Sethi AA, Remaley AT, Schnohr P, Grande P, Tybjaerg-Hansen A. Association of loss-of-function mutations in the ABCA1 gene with high-density lipoprotein cholesterol levels and risk of ischemic heart disease. JAMA. 2008;299(21):2524–32. doi: 10.1001/jama.299.21.2524. http://dx.doi.org/10.1001/jama.299.21.2524. [DOI] [PubMed] [Google Scholar]
  • 9.Voight BF, Peloso GM, Orho-Melander M, Frikke-Schmidt R, Barbalic M, Jensen MK, Hindy G, Hólm H, Ding EL, Johnson T, Schunkert H, Samani NJ, Clarke R, Hopewell JC, Thompson JF, Li M, Thorleifsson G, Newton-Cheh C, Musunuru K, Pirruccello JP, Saleheen D, Chen L, Stewart A, Schillert A, Thorsteinsdottir U, Thorgeirsson G, Anand S, Engert JC, Morgan T, Spertus J, Stoll M, Berger K, Martinelli N, Girelli D, McKeown PP, Patterson CC, Epstein SE, Devaney J, Burnett MS, Mooser V, Ripatti S, Surakka I, Nieminen MS, Sinisalo J, Lokki ML, Perola M, Havulinna A, de Faire U, Gigante B, Ingelsson E, Zeller T, Wild P, de Bakker PI, Klungel OH, Maitland-van der Zee AH, Peters BJ, de Boer A, Grobbee DE, Kamphuisen PW, Deneer VH, Elbers CC, Onland-Moret NC, Hofker MH, Wijmenga C, Verschuren WM, Boer JM, van der Schouw YT, Rasheed A, Frossard P, Demissie S, Willer C, Do R, Ordovas JM, Abecasis GR, Boehnke M, Mohlke KL, Daly MJ, Guiducci C, Burtt NP, Surti A, Gonzalez E, Purcell S, Gabriel S, Marrugat J, Peden J, Erdmann J, Diemert P, Willenborg C, König IR, Fischer M, Hengstenberg C, Ziegler A, Buysschaert I, Lambrechts D, Van de Werf F, Fox KA, El Mokhtari NE, Rubin D, Schrezenmeir J, Schreiber S, Schäfer A, Danesh J, Blankenberg S, Roberts R, McPherson R, Watkins H, Hall AS, Overvad K, Rimm E, Boerwinkle E, Tybjaerg-Hansen A, Cupples LA, Reilly MP, Melander O, Mannucci PM, Ardissino D, Siscovick D, Elosua R, Stefansson K, O’Donnell CJ, Salomaa V, Rader DJ, Peltonen L, Schwartz SM, Altshuler D, Kathiresan S. Plasma HDL cholesterol and risk of myocardial infarction: a mendelian randomisation study. Lancet. 2012;380(9841):572–80. doi: 10.1016/S0140-6736(12)60312-2. http://dx.doi.org/10.1016/S0140-6736(12)60312-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.van Capelleveen JC, Bochem AE, Motazacker MM, Hovingh GK, Kastelein JJ. Genetics of HDL-C: a causal link to atherosclerosis? Curr Atheroscler Rep. 2013;15(6):326. doi: 10.1007/s11883-013-0326-8. http://dx.doi.org/10.1007/s11883-013-0326-8. [DOI] [PubMed] [Google Scholar]
  • 11.Angeloni E, Paneni F, Landmesser U, Benedetto U, Melina G, Lüscher TF, Volpe M, Sinatra R, Cosentino F. Lack of protective role of HDL-C in patients with coronary artery disease undergoing elective coronary artery bypass grafting. Eur Heart J. 2013;34(46):3557–62. doi: 10.1093/eurheartj/eht163. http://dx.doi.org/10.1093/eurheartj/eht163. [DOI] [PubMed] [Google Scholar]
  • 12.van der Steeg WA, Holme I, Boekholdt SM, Larsen ML, Lindahl C, Stroes ES, Tikkanen MJ, Wareham NJ, Faergeman O, Olsson AG, Pedersen TR, Khaw KT, Kastelein JJ. High-density lipoprotein cholesterol, high-density lipoprotein particle size, and apolipoprotein A-I: significance for cardiovascular risk: the IDEAL and EPIC-Norfolk studies. J Am Coll Cardiol. 2008;51(6):634–42. doi: 10.1016/j.jacc.2007.09.060. http://dx.doi.org/10.1016/j.jacc.2007.09.060. [DOI] [PubMed] [Google Scholar]
  • 13.Barter PJ, Caulfield M, Eriksson M, Grundy SM, Kastelein JJ, Komajda M, Lopez-Sendon J, Mosca L, Tardif JC, Waters DD, Shear CL, Revkin JH, Buhr KA, Fisher MR, Tall AR, Brewer B ILLUMINATE Investigators. Effects of torcetrapib in patients at high risk for coronary events. N Engl J Med. 2007;357(21):2109–22. doi: 10.1056/NEJMoa0706628. http://dx.doi.org/10.1056/NEJMoa0706628. [DOI] [PubMed] [Google Scholar]
  • 14.Schwartz GG, Olsson AG, Abt M, Ballantyne CM, Barter PJ, Brumm J, Chaitman BR, Holme IM, Kallend D, Leiter LA, Leitersdorf E, McMurray JJ, Mundl H, Nicholls SJ, Shah PK, Tardif JC, Wright RS dal-OUTCOMES Investigators. Effects of dalcetrapib in patients with a recent acute coronary syndrome. N Engl J Med. 2012;367(22):2089–99. doi: 10.1056/NEJMoa1206797. http://dx.doi.org/10.1056/NEJMoa1206797. [DOI] [PubMed] [Google Scholar]
  • 15.Lincoff AM, Nicholls SJ, Riesmeyer JS, Barter PJ, Brewer HB, Fox KAA, Gibson CM, Granger C, Menon V, Montalescot G, Rader D, Tall AR, McErlean E, Wolski K, Ruotolo G, Vangerow B, Weerakkody G, Goodman SG, Conde D, McGuire DK, Nicolau JC, Leiva-Pons JL, Pesant Y, Li W, Kandath D, Kouz S, Tahirkheli N, Mason D, Nissen SE ACCELERATE Investigators. Evacetrapib and cardiovascular outcomes in high-risk vascular disease. N Engl J Med. 2017;376(20):1933–42. doi: 10.1056/NEJMoa1609581. http://dx.doi.org/10.1056/NEJMoa1609581. [DOI] [PubMed] [Google Scholar]
  • 16.HPS3/TIMI55–REVEAL Collaborative Group. Bowman L, Hopewell JC, Chen F, Wallendszus K, Stevens W, Collins R, Wiviott SD, Cannon CP, Braunwald E, Sammons E, Landray MJ. Effects of anacetrapib in patients with atherosclerotic vascular disease. N Engl J Med. 2017;377(13):1217–27. doi: 10.1056/NEJMoa1706444. http://dx.doi.org/10.1056/NEJMoa1706444. [DOI] [PubMed] [Google Scholar]
  • 17.Tailleux A, Fruchart JC. HDL heterogeneity and atherosclerosis. Crit Rev Clin Lab Sci. 1996;33(3):163–201. doi: 10.3109/10408369609083060. http://dx.doi.org/10.3109/10408369609083060. [DOI] [PubMed] [Google Scholar]
  • 18.Rader DJ. Molecular regulation of HDL metabolism and function: implications for novel therapies. J Clin Invest. 2006;116(12):3090–100. doi: 10.1172/JCI30163. http://dx.doi.org/10.1172/JCI30163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Vaisar T, Pennathur S, Green PS, Gharib SA, Hoofnagle AN, Cheung MC, Byun J, Vuletic S, Kassim S, Singh P, Chea H, Knopp RH, Brunzell J, Geary R, Chait A, Zhao XQ, Elkon K, Marcovina S, Ridker P, Oram JF, Heinecke JW. Shotgun proteomics implicates protease inhibition and complement activation in the antiinflammatory properties of HDL. J Clin Invest. 2007;117(3):746–56. doi: 10.1172/JCI26206. http://dx.doi.org/10.1172/JCI26206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kontush A, Lhomme M, Chapman MJ. Unraveling the complexities of the HDL lipidome. J Lipid Res. 2013;54(11):2950–63. doi: 10.1194/jlr.R036095. http://dx.doi.org/10.1194/jlr.R036095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Rosenson RS, Brewer HB, Jr, Chapman MJ, Fazio S, Hussain MM, Kontush A, Krauss RM, Otvos JD, Remaley AT, Schaefer EJ. HDL measures, particle heterogeneity, proposed nomenclature, and relation to atherosclerotic cardiovascular events. Clin Chem. 2011;57(3):392–410. doi: 10.1373/clinchem.2010.155333. http://dx.doi.org/10.1373/clinchem.2010.155333. [DOI] [PubMed] [Google Scholar]
  • 22.Camont L, Chapman MJ, Kontush A. Biological activities of HDL subpopulations and their relevance to cardiovascular disease. Trends Mol Med. 2011;17(10):594–603. doi: 10.1016/j.molmed.2011.05.013. http://dx.doi.org/10.1016/j.molmed.2011.05.013. [DOI] [PubMed] [Google Scholar]
  • 23.Toth PP. Reverse cholesterol transport: high-density lipoprotein’s magnificent mile. Curr Atheroscler Rep. 2003;5(5):386–93. doi: 10.1007/s11883-003-0010-5. http://dx.doi.org/10.1007/s11883-003-0010-5. [DOI] [PubMed] [Google Scholar]
  • 24.Kontush A, Chapman MJ. Antiatherogenic function of HDL particle subpopulations: focus on antioxidative activities. Curr Opin Lipidol. 2010;21(4):312–8. doi: 10.1097/MOL.0b013e32833bcdc1. http://dx.doi.org/10.1097/MOL.0b013e32833bcdc1. [DOI] [PubMed] [Google Scholar]
  • 25.Barter PJ, Nicholls S, Rye KA, Anantharamaiah GM, Navab M, Fogelman AM. Antiinflammatory properties of HDL. Circ Res. 2004;95(8):764–72. doi: 10.1161/01.RES.0000146094.59640.13. http://dx.doi.org/10.1161/01.RES.0000146094.59640.13. [DOI] [PubMed] [Google Scholar]
  • 26.Mineo C, Deguchi H, Griffin JH, Shaul PW. Endothelial and antithrombotic actions of HDL. Circ Res. 2006;98(11):1352–64. doi: 10.1161/01.RES.0000225982.01988.93. http://dx.doi.org/10.1161/01.RES.0000225982.01988.93. [DOI] [PubMed] [Google Scholar]
  • 27.Sugano M, Tsuchida K, Makino N. High-density lipoproteins protect endothelial cells from tumor necrosis factor-alpha-induced apoptosis. Biochem Biophys Res Commun. 2000;272(3):872–6. doi: 10.1006/bbrc.2000.2877. http://dx.doi.org/10.1006/bbrc.2000.2877. [DOI] [PubMed] [Google Scholar]
  • 28.Gualandri V, Franceschini G, Sirtori CR, Gianfranceschi G, Orsini GB, Cerrone A, Menotti A. AIMilano apoprotein identification of the complete kindred and evidence of a dominant genetic transmission. Am J Hum Genet. 1985;37(6):1083–97. [PMC free article] [PubMed] [Google Scholar]
  • 29.Sirtori CR, Calabresi L, Franceschini G, Baldassarre D, Amato M, Johansson J, Salvetti M, Monteduro C, Zulli R, Muiesan ML, Agabiti-Rosei E. Cardiovascular status of carriers of the apolipoprotein A-I(Milano) mutant: the Limone sul Garda study. Circulation. 2001;103(15):1949–54. doi: 10.1161/01.cir.103.15.1949. http://dx.doi.org/10.1161/01.CIR.103.15.1949. [DOI] [PubMed] [Google Scholar]
  • 30.Nissen SE, Tsunoda T, Tuzcu EM, Schoenhagen P, Cooper CJ, Yasin M, Eaton GM, Lauer MA, Sheldon WS, Grines CL, Halpern S, Crowe T, Blankenship JC, Kerensky R. Effect of recombinant ApoA-I Milano on coronary atherosclerosis in patients with acute coronary syndromes: a randomized controlled trial. JAMA. 2003;290(17):2292–300. doi: 10.1001/jama.290.17.2292. http://dx.doi.org/10.1001/jama.290.17.2292. [DOI] [PubMed] [Google Scholar]
  • 31.Ibanez B, Giannarelli C, Cimmino G, Santos-Gallego CG, Alique M, Pinero A, Vilahur G, Fuster V, Badimon L, Badimon JJ. Recombinant HDL(Milano) exerts greater anti-inflammatory and plaque stabilizing properties than HDL(wild-type) Atherosclerosis. 2012;220(1):72–7. doi: 10.1016/j.atherosclerosis.2011.10.006. http://dx.doi.org/10.1016/j.atherosclerosis.2011.10.006. [DOI] [PubMed] [Google Scholar]
  • 32.Chyu KY, Shah PK. HDL/ApoA-1 infusion and ApoA-1 gene therapy in atherosclerosis. Front Pharmacol. 2015;6:187. doi: 10.3389/fphar.2015.00187. http://dx.doi.org/10.3389/fphar.2015.00187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Salonen JT, Salonen R, Seppänen K, Rauramaa R, Tuomilehto J. HDL, HDL2, and HDL3 subfractions, and the risk of acute myocardial infarction. A prospective population study in eastern Finnish men. Circulation. 1991;84(1):129–39. doi: 10.1161/01.cir.84.1.129. http://dx.doi.org/10.1161/01.CIR.84.1.129. [DOI] [PubMed] [Google Scholar]
  • 34.Drexel H, Amann FW, Rentsch K, Neuenschwander C, Luethy A, Khan SI, Follath F. Relation of the level of high-density lipoprotein subfractions to the presence and extent of coronary artery disease. Am J Cardiol. 1992;70(4):436–40. doi: 10.1016/0002-9149(92)91186-8. http://dx.doi.org/10.1016/0002-9149(92)91186-8. [DOI] [PubMed] [Google Scholar]
  • 35.Freedman DS, Otvos JD, Jeyarajah EJ, Barboriak JJ, Anderson AJ, Walker JA. Relation of lipoprotein subclasses as measured by proton nuclear magnetic resonance spectroscopy to coronary artery disease. Arterioscler Thromb Vasc Biol. 1998;18(7):1046–53. doi: 10.1161/01.atv.18.7.1046. http://dx.doi.org/10.1161/01.ATV.18.7.1046. [DOI] [PubMed] [Google Scholar]
  • 36.Rosenson RS, Otvos JD, Freedman DS. Relations of lipoprotein subclass levels and low-density lipoprotein size to progression of coronary artery disease in the Pravastatin Limitation of Atherosclerosis in the Coronary Arteries (PLAC-I) trial. Am J Cardiol. 2002;90(2):89–94. doi: 10.1016/s0002-9149(02)02427-x. http://doi.org/10.1016/S0002-9149(02)02427-X. [DOI] [PubMed] [Google Scholar]
  • 37.AIM-HIGH Investigators. Boden WE, Probstfield JL, Anderson T, Chaitman BR, Desvignes-Nickens P, Koprowicz K, McBride R, Teo K, Weintraub W. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med. 2011;365(24):2255–67. doi: 10.1056/NEJMoa1107579. http://dx.doi.org/10.1056/NEJMoa1107579. [DOI] [PubMed] [Google Scholar]
  • 38.HPS2-THRIVE Collaborative Group. Landray MJ, Haynes R, Hopewell JC, Parish S, Aung T, Tomson J, Wallendszus K, Craig M, Jiang L, Collins R, Armitage J. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med. 2014;371(3):203–12. doi: 10.1056/NEJMoa1300955. http://dx.doi.org/10.1056/NEJMoa1300955. [DOI] [PubMed] [Google Scholar]
  • 39.Kontush A. HDL particle number and size as predictors of cardiovascular disease. Front Pharmacol. 2015;6:218. doi: 10.3389/fphar.2015.00218. http://dx.doi.org/10.3389/fphar.2015.00218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ishigami M, Yamashita S, Sakai N, Arai T, Hirano K, Hiraoka H, Kameda-Takemura K, Matsuzawa Y. Large and cholesteryl ester-rich high-density lipoproteins in cholesteryl ester transfer protein (CETP) deficiency can not protect macrophages from cholesterol accumulation induced by acetylated low-density lipoproteins. J Biochem. 1994;116(2):257–62. doi: 10.1093/oxfordjournals.jbchem.a124516. http://dx.doi.org/10.1093/oxfordjournals.jbchem.a124516. [DOI] [PubMed] [Google Scholar]
  • 41.Qi Y, Fan J, Liu J, Wang W, Wang M, Sun J, Liu J, Xie W, Zhao F, Li Y, Zhao D. Cholesterol-overloaded HDL particles are independently associated with progression of carotid atherosclerosis in a cardiovascular disease-free population: a community-based cohort study. J Am Coll Cardiol. 2015;65(4):355–63. doi: 10.1016/j.jacc.2014.11.019. http://dx.doi.org/10.1016/j.jacc.2014.11.019. [DOI] [PubMed] [Google Scholar]
  • 42.Kontush A, Chantepie S, Chapman MJ. Small, dense HDL particles exert potent protection of atherogenic LDL against oxidative stress. Arterioscler Thromb Vasc Biol. 2003;23(10):1881–8. doi: 10.1161/01.ATV.0000091338.93223.E8. http://dx.doi.org/10.1161/01.ATV.0000091338.93223.E8. [DOI] [PubMed] [Google Scholar]
  • 43.Parish S, Offer A, Clarke R, Hopewell JC, Hill MR, Otvos JD, Armitage J, Collins R Heart Protection Study Collaborative Group. Lipids and lipoproteins and risk of different vascular events in the MRC/BHF Heart Protection Study. Circulation. 2012;125(20):2469–78. doi: 10.1161/CIRCULATIONAHA.111.073684. http://dx.doi.org/10.1161/CIRCULATIONAHA.111.073684. [DOI] [PubMed] [Google Scholar]
  • 44.Mackey RH, Greenland P, Goff DC, Jr, Lloyd-Jones D, Sibley CT, Mora S. High-density lipoprotein cholesterol and particle concentrations, carotid atherosclerosis, and coronary events: MESA (multi-ethnic study of atherosclerosis) J Am Coll Cardiol. 2012;60(6):508–16. doi: 10.1016/j.jacc.2012.03.060. http://dx.doi.org/10.1016/j.jacc.2012.03.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Van Lenten BJ, Navab M, Shih D, Fogelman AM, Lusis AJ. The role of high-density lipoproteins in oxidation and inflammation. Trends Cardiovasc Med. 2001;11(3–4):155–61. doi: 10.1016/s1050-1738(01)00095-0. [DOI] [PubMed] [Google Scholar]
  • 46.Navab M, Reddy ST, Van Lenten BJ, Fogelman AM. HDL and cardiovascular disease: atherogenic and atheroprotective mechanisms. Nat Rev Cardiol. 2011;8(4):222–32. doi: 10.1038/nrcardio.2010.222. http://dx.doi.org/10.1038/nrcardio.2010.222. [DOI] [PubMed] [Google Scholar]
  • 47.Van Lenten BJ, Hama SY, de Beer FC, Stafforini DM, McIntyre TM, Prescott SM, La Du BN, Fogelman AM, Navab M. Anti-inflammatory HDL becomes pro-inflammatory during the acute phase response. Loss of protective effect of HDL against LDL oxidation in aortic wall cell cocultures. J Clin Invest. 1995;96(6):2758–67. doi: 10.1172/JCI118345. http://dx.doi.org/10.1172/JCI118345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Huang Y, DiDonato JA, Levison BS, Schmitt D, Li L, Wu Y, Buffa J, Kim T, Gerstenecker GS, Gu X, Kadiyala CS, Wang Z, Culley MK, Hazen JE, Didonato AJ, Fu X, Berisha SZ, Peng D, Nguyen TT, Liang S, Chuang CC, Cho L, Plow EF, Fox PL, Gogonea V, Tang WH, Parks JS, Fisher EA, Smith JD, Hazen SL. An abundant dysfunctional apolipoprotein A1 in human atheroma. Nat Med. 2014;20(2):193–203. doi: 10.1038/nm.3459. http://dx.doi.org/10.1038/nm.3459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Carnuta MG, Stancu CS, Toma L, Sanda GM, Niculescu LS, Deleanu M, Popescu AC, Popescu MR, Vlad A, Dimulescu DR, Simionescu M, Sima AV. Dysfunctional high-density lipoproteins have distinct composition, diminished anti-inflammatory potential and discriminate acute coronary syndrome from stable coronary artery disease patients. Sci Rep. 2017;7(1):7295. doi: 10.1038/s41598-017-07821-5. http://dx.doi.org/10.1038/s41598-017-07821-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hedrick CC, Thorpe SR, Fu MX, Harper CM, Yoo J, Kim SM, Wong H, Peters AL. Glycation impairs high-density lipoprotein function. Diabetologia. 2000;43(3):312–20. doi: 10.1007/s001250050049. http://dx.doi.org/10.1007/s001250050049. [DOI] [PubMed] [Google Scholar]
  • 51.Hansel B, Giral P, Nobecourt E, Chantepie S, Bruckert E, Chapman MJ, Kontush A. Metabolic syndrome is associated with elevated oxidative stress and dysfunctional dense high-density lipoprotein particles displaying impaired antioxidative activity. J Clin Endocrinol Metab. 2004;89(10):4963–71. doi: 10.1210/jc.2004-0305. http://dx.doi.org/10.1210/jc.2004-0305. [DOI] [PubMed] [Google Scholar]
  • 52.Holzer M, Gauster M, Pfeifer T, Wadsack C, Fauler G, Stiegler P, Koefeler H, Beubler E, Schuligoi R, Heinemann A, Marsche G. Protein carbamylation renders high-density lipoprotein dysfunctional. Antioxid Redox Signal. 2011;14(12):2337–46. doi: 10.1089/ars.2010.3640. http://dx.doi.org/10.1089/ars.2010.3640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Fisher EA, Feig JE, Hewing B, Hazen SL, Smith JD. High-density lipoprotein function, dysfunction, and reverse cholesterol transport. Arterioscler Thromb Vasc Biol. 2012;32(12):2813–20. doi: 10.1161/ATVBAHA.112.300133. http://dx.doi.org/10.1161/ATVBAHA.112.300133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Woudberg NJ, Goedecke JH, Blackhurst D, Frias M, James R, Opie LH, Lecour S. Association between ethnicity and obesity with high-density lipoprotein (HDL) function and subclass distribution. Lipids Health Dis. 2016;15:92. doi: 10.1186/s12944-016-0257-9. http://dx.doi.org/10.1186/s12944-016-0257-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.McMorrow AM, O’Reilly M, Connaughton RM, Carolan E, O’Shea D, Lithander FE, McGillicuddy FC, Roche HM. Obesity and dietary fat modulate HDL function in adolescents: results from a cross-sectional analysis and a randomized, placebo-controlled, crossover trial. FASEB J. 2016;30(1 Suppl) Abstract #130.6. [Google Scholar]
  • 56.Zvintzou E, Skroubis G, Chroni A, Petropoulou PI, Gkolfinopoulou C, Sakellaropoulos G, Gantz D, Mihou I, Kalfarentzos F, Kypreos KE. Effects of bariatric surgery on HDL structure and functionality: results from a prospective trial. J Clin Lipidol. 2014;8(4):408–17. doi: 10.1016/j.jacl.2014.05.001. http://dx.doi.org/10.1016/j.jacl.2014.05.001. [DOI] [PubMed] [Google Scholar]
  • 57.Nicholls SJ, Lundman P, Harmer JA, Cutri B, Griffiths KA, Rye KA, Barter PJ, Celermajer DS. Consumption of saturated fat impairs the anti-inflammatory properties of high-density lipoproteins and endothelial function. J Am Coll Cardiol. 2006;48(4):715–20. doi: 10.1016/j.jacc.2006.04.080. http://dx.doi.org/10.1016/j.jacc.2006.04.080. [DOI] [PubMed] [Google Scholar]
  • 58.Adorni MP, Zimetti F, Billheimer JT, Wang N, Rader DJ, Phillips MC, Rothblat GH. The roles of different pathways in the release of cholesterol from macrophages. J Lipid Res. 2007;48(11):2453–62. doi: 10.1194/jlr.M700274-JLR200. http://dx.doi.org/10.1194/jlr.M700274-JLR200. [DOI] [PubMed] [Google Scholar]
  • 59.Cuchel M, Rader DJ. Macrophage reverse cholesterol transport: key to the regression of atherosclerosis? Circulation. 2006;113(21):2548–55. doi: 10.1161/CIRCULATIONAHA.104.475715. http://dx.doi.org/10.1161/CIRCULATIONAHA.104.475715. [DOI] [PubMed] [Google Scholar]
  • 60.Khera AV, Cuchel M, de la Llera-Moya M, Rodrigues A, Burke MF, Jafri K, French BC, Phillips JA, Mucksavage ML, Wilensky RL, Mohler ER, Rothblat GH, Rader DJ. Cholesterol efflux capacity, high-density lipoprotein function, and atherosclerosis. N Engl J Med. 2011;364(2):127–35. doi: 10.1056/NEJMoa1001689. http://dx.doi.org/10.1056/NEJMoa1001689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Rohatgi A, Khera A, Berry JD, Givens EG, Ayers CR, Wedin KE, Neeland IJ, Yuhanna IS, Rader DR, de Lemos JA, Shaul PW. HDL cholesterol efflux capacity and incident cardiovascular events. N Engl J Med. 2014;371(25):2383–93. doi: 10.1056/NEJMoa1409065. http://dx.doi.org/10.1056/NEJMoa1409065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Rohatgi A. High-Density lipoprotein function measurement in human studies: focus on cholesterol efflux capacity. Prog Cardiovasc Dis. 2015;58(1):32–40. doi: 10.1016/j.pcad.2015.05.004. http://dx.doi.org/10.1016/j.pcad.2015.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.MDCO-216 infusions leading to changes in atherosclerosis: a novel therapy in development to improve cardiovascular outcomes – proof of concept intravascular ultrasound (IVUS), lipids, and other surrogate biomarkers trial (PILOT) ClinicalTrials.gov Identifier: NCT02678923. [Last accessed 13 July 2017]. Available at http://clinicaltrials.gov/ct2/show/results/NCT02678923.
  • 64.Waksman R, Torguson R, Kent KM, Pichard AD, Suddath WO, Satler LF, Martin BD, Perlman TJ, Maltais JA, Weissman NJ, Fitzgerald PJ, Brewer HB., Jr A first-in-man, randomized, placebo-controlled study to evaluate the safety and feasibility of autologous delipidated high-density lipoprotein plasma infusions in patients with acute coronary syndrome. J Am Coll Cardiol. 2010;55(24):2727–35. doi: 10.1016/j.jacc.2009.12.067. http://dx.doi.org/10.1016/j.jacc.2009.12.067. [DOI] [PubMed] [Google Scholar]
  • 65.Michael Gibson C, Korjian S, Tricoci P, Daaboul Y, Yee M, Jain P, Alexander JH, Steg PG, Lincoff AM, Kastelein JJ, Mehran R, D’Andrea DM, Deckelbaum LI, Merkely B, Zarebinski M, Ophuis TO, Harrington RA. Safety and tolerability of CSL112, a reconstituted, infusible, plasma-derived apolipoprotein A-I, after acute myocardial infarction: the AEGIS-I trial (ApoA-I event reducing in ischemic syndromes I) Circulation. 2016;134(24):1918–30. doi: 10.1161/CIRCULATIONAHA.116.025687. http://dx.doi.org/10.1161/CIRCULATIONAHA.116.025687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Asokan A, Samulski RJ. An emerging adeno-associated viral vector pipeline for cardiac gene therapy. Hum Gene Ther. 2013;24(11):906–13. doi: 10.1089/hum.2013.2515. http://dx.doi.org/10.1089/hum.2013.2515. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Drugs in Context are provided here courtesy of BioExcel Publishing Ltd

RESOURCES